Language selection

Search

Patent 2989831 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2989831
(54) English Title: CRISPR/CAS9 COMPLEX FOR GENOMIC EDITING
(54) French Title: COMPLEXE CRISPR/CAS9 POUR L'EDITION GENOMIQUE
Status: Deemed Abandoned and Beyond the Period of Reinstatement - Pending Response to Notice of Disregarded Communication
Bibliographic Data
(51) International Patent Classification (IPC):
  • C12Q 1/68 (2018.01)
  • A61K 48/00 (2006.01)
  • C12N 9/22 (2006.01)
  • C12N 15/00 (2006.01)
  • C12N 15/113 (2010.01)
  • C12N 15/90 (2006.01)
(72) Inventors :
  • TOWNES, TIM (United States of America)
  • DING, LEI (United States of America)
  • CHANG, CHIA-WEI (United States of America)
(73) Owners :
  • THE UAB RESEARCH FOUNDATION
(71) Applicants :
  • THE UAB RESEARCH FOUNDATION (United States of America)
(74) Agent: MARKS & CLERK
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2016-06-17
(87) Open to Public Inspection: 2016-12-22
Availability of licence: N/A
Dedicated to the Public: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2016/038161
(87) International Publication Number: US2016038161
(85) National Entry: 2017-12-15

(30) Application Priority Data:
Application No. Country/Territory Date
62/181,138 (United States of America) 2015-06-17
62/266,316 (United States of America) 2015-12-11

Abstracts

English Abstract

Provided herein are CRISPR/Cas9 complexes and method of using same.


French Abstract

La présente invention concerne des complexes CRISPR/Cas9 et un procédé d'utilisation de ces complexes.

Claims

Note: Claims are shown in the official language in which they were submitted.


What is claimed is:
1. A complex for correcting a mutation in the genome of a cell comprising
a. a guide RNA (gRNA) comprising a first nucleotide sequence that hybridizes
to a target DNA in the genome of a cell, wherein the target DNA comprises a
mutation, and a second nucleotide sequence that interacts with a site-directed
nuclease;
b. a recombinant site-directed nuclease operably linked to a supercharged
protein, wherein the site-directed nuclease comprises an RNA-binding portion
that interacts with the second nucleotide sequence of the guide RNA and
wherein the site-directed nuclease specifically binds and cleaves the target
DNA to create a double stranded break; and
c. a single-stranded donor oligonucleotide (ssODN) that hybridizes to a
genomic
sequence flanking the double stranded break in the target DNA and integrates
into the target DNA to correct a mutation in the target DNA.
2. The complex of claim 1, wherein the supercharged protein is operably
linked to the
amino-terminus or the carboxy-terminus of the nuclease.
3. The complex of any of claims 1 or 2, wherein the complex further
comprises a trans-
activating transcriptional activator (TAT) peptide that is operably linked to
the amino-
terminus of the site-directed nuclease.
4. The complex of any of claims 1-3, wherein the complex further comprises
a trans-
activating transcriptional activator (TAT) peptide that is operably linked to
the
carboxy-terminus of the site-directed nuclease.
5. The complex of any of claims 1-4, wherein the complex further comprises a
negatively charged peptide of about 10 to about 25 amino acids in length that
is
operably linked to the carboxy-terminus of the site-directed nuclease.
6. The complex of claim 5, wherein the negatively charged peptide is an INF7
peptide
comprising SEQ ID NO: 50.

7. The complex of any of claims 1-6, wherein the supercharged protein has
an overall
positive charge that is greater than its corresponding unmodified protein.
8. The complex of claim 7, wherein the overall positive charge is from about
+5 to about
+40.
9. The complex of claim 8, wherein the supercharged protein is superpositively
charged
green fluorescent protein (GFP).
10. The complex of claim 9, wherein the supercharged protein is a
superpositively
charged +36 GFP.
11. The complex of any of claims 1-10, wherein the ssODN that hybridizes to
the
genomic sequence flanking the double stranded break in the target DNA is a
template
for homology directed repair of a mutation in the target DNA.
12. The complex of claim 11, wherein the ssODN hybridizes to the genomic
sequence
encoding hemoglobin.
13. The complex of any of claims 1-12, wherein the nuclease is Cas9.
14. The complex of any of claims 1-13, wherein the molar ratio of gRNA to site-
directed
nuclease operably linked to a supercharged protein to ssODN is from about
1:1:0.2 to
about 1.5:1:2Ø
15. The complex of any of claims 1-14, wherein the molar ratio of gRNA to site-
directed
nuclease operably linked to a supercharged protein to ssODN is from about
1:1:1 to
about 1.5:1:1:15.
16. A cell comprising the complex of any of claims 1-15.
17. The cell of claim 16, wherein the cell is a eukaryotic cell.
56

18. The cell of claim 17, wherein the eukaryotic cell is a human cell.
19. The cell of any of claims 16-18, wherein the cell is a germ cell, a stem
cell, or a
precursor cell.
20. The cell of claim 19, wherein the stem cell is an induced pluripotent stem
cell.
21. The cell of claim 19, wherein the precursor cell is a hematopoietic stem
cell.
22. The cell of any of claims 16-21, wherein the cell is in vitro, ex vivo or
in vivo.
23. A method of site-specific modification of a target DNA in a population of
cells
comprising introducing into the cells the complex of any of claims 1-15,
wherein the
complex is introduced into the cells under conditions that allow homology-
directed
repair (HDR) and integration of the ssODN into the target DNA.
24. The method of claim 23, wherein the complex is introduced into the cells
by
nucleoporation.
25. The method of claim 23 or 24, wherein the ssODN integrated into the target
DNA
corrects a mutation in the target DNA.
26. The method of any of claims 23-25, wherein the ratio of homology-directed
repair to
nonhomologous end joining (NHEJ) in the population of cells is from about 10
to
about 0.5.
27. The method of any of claims 23-26, wherein the mutation is corrected in at
least 5%
of the cells.
28. The method of claim 27, wherein the the cell survival rate for corrected
cells is at
least about 50%.
57

29. The method of any of claims 23-28, wherein the molar ratio of gRNA to site-
directed
nuclease operably linked to a supercharged protein to ssODN is from about
1:1:0.2 to
about 1.5:1:2Ø
30. The method of any of claims 23-29, wherein the target DNA encodes
hemoglobin.
31. The method of claim 30, wherein the site-specific modification corrects a
hemoglobin
mutation associated with sickle cell anemia.
32. The method of claim 30, wherein the site-specific modification corrects a
mutation
associated with .beta.-thalassemia.
33. A method of treating a disease associated with a mutation in the genomic
sequence
encoding hemoglobin in a subject comprising:
a. introducing into a population of cells obtained from the subject the
complex of
claim 11 under conditions that allow homology-directed repair (HDR) to
correct the mutation in the genomic sequence encoding hemoglobin and
b. transplanting the cells of step (a) into the subject.
34. The method of claim 33, wherein the disease associated with a mutation in
the
genomic sequence encoding hemoglobin is sickle cell disease or .beta.-
thalassemia.
35. The method of claim 33 or 34, wherein the cell is a hematopoietic stem
cell or an
induced pluripotent stem cell.
36. The method of claim any of claims 33-35, wherein at least 5% of the
transplanted
cells include a corrected mutation.
37. The method of any of claims 33-36, wherein the ratio of homology-directed
repair to
nonhomologous end joining in the population of cells is at least about 0.5.
38. The method of any of claims 33-36, wherein the complex is introduced into
the cell
by nucleoporation.
58

39. A method of correcting a mutation associated with a T-cell disorder
comprising
introducing into a population of cells obtained from a subject with the T-cell
disorder
a complex comprising:
a. a guide RNA (gRNA) comprising a first nucleotide sequence that hybridizes
to a target DNA in the genome of a cell, wherein the target DNA comprises
the mutation associated with the T-cell disorder, and a second nucleotide
sequence that interacts with a site-directed nuclease;
b. a recombinant site-directed nuclease operably linked to a supercharged
protein, wherein the site-directed nuclease comprises an RNA-binding portion
that interacts with the second nucleotide sequence of the gRNA and wherein
the site-directed nuclease specifically binds and cleaves the target DNA that
comprises the mutation associated with the T-cell disorder to create a double
stranded break in the target DNA; and
c. a single stranded donor oligonucleotide (ssODN) comprising a third
nucleotide sequence that hybridizes to a genomic sequence flanking the double
stranded break in the target DNA and that integrates into the target DNA to
correct the mutation associated with the T-cell disorder,
wherein the complex is introduced into the cell under conditions that allow
homology-
directed repair (HDR) to correct the mutation associated with the T-cell
disorder.
40. The method of claim 39, wherein the target DNA encodes a protein
associated with T-
lymphocyte development.
41. The method of claim 39 or 40, wherein the cells are selected from the
group
consisting of hematopoietic stem cells or pluripotent stem cells.
42. The method of claim 41, wherein the pluripotent stem cells are induced
pluripotent
stem cells.
43. The method of any of claims 39-42, wherein the supercharged protein is
operably
linked to the amino-terminus or the carboxy-terminus of the nuclease.
44. The method of any of claims 39-43, wherein the recombinant site-directed
nuclease
operably linked to a supercharged protein further comprises a trans-activating
59

transcriptional activator (TAT) peptide operably linked to the amino-terminus
of the
site-directed nuclease.
45. The method of any of claims 39-44, wherein the complex further comprises a
trans-
activating transcriptional activator (TAT) peptide that is operably linked to
the
carboxy-terminus of the site-directed nuclease.
46. The method of any of claims 39-45 wherein the complex further comprises a
negatively charged peptide of about 10 to about 25 amino acids in length that
is
operably linked to the carboxy-terminus of the site-directed nuclease.
47. The method of claim 46, wherein the negatively charged peptide is an INF7
peptide
comprising SEQ ID NO: 50.
48. The method of any of claims 39-47, wherein the supercharged protein has an
overall
positive charge that is greater than its corresponding unmodified protein.
49. The method of claim 48, wherein the overall positive charge is from about
+5 to about
about +40.
50. The method of any of claims 39-49, wherein the supercharged protein is
superpositively charged green fluorescent protein (GFP).
51. The method of claim 50, wherein the supercharged protein is a
superpositively
charged +36 GFP.
52. The method of any of claims 39-51, wherein the nuclease is Cas9.
53. The method of any of claims 39-52, wherein the molar ratio of gRNA to site-
directed
nuclease operably linked to a supercharged protein to ssODN is from about
1:1:0.2 to
about 1.5:1:2Ø

54. The method of any of claims 39-53, wherein the ratio of homology-directed
repair to
nonhomologous end joining in the population of cells obtained from a subject
with the
T-cell disorder is at least about 0.5.
55. The method of any of claims 39-54, wherein the complex is introduced into
the cells
by nucleoporation.
56. The method of any of claims 39-55, wherein at least 5% of the population
of cells
obtained from a subject with the T-cell disorder undergo HDR to correct the
mutation
associated with the T-cell disorder.
57. The method of any of claims 39-56, further comprising isolating the cells
corrected
with HDR.
58. The method of claim 57, further comprising culturing the cells corrected
with HDR.
59. The method of claim 58, wherein the cells are cultured under conditions
for
expansion.
60. The method of claim 58 or 59, wherein the cells are cultured under
conditions that
promote differentiation of the cells into T cells.
61. A method of treating a T cell disorder associated with a genetic mutation
in a subject
comprising transplanting into the subject cells obtained by the method of any
one of
claims 39-60.
62. The method of claim 61, wherein the transplantation is autologous.
63. The method of any of claims 39-62, wherein the T cell disorder is severe
combined
immune deficiency.
64. A complex for correcting a mutation associated with a T-cell disorder in
the genome
of a cell comprising:
61

a. a guide RNA (gRNA) comprising a first nucleotide sequence that hybridizes
to a target DNA in the genome of a cell, wherein the target DNA comprises
the mutation associated with the T-cell disorder, and a second nucleotide
sequence that interacts with a site-directed nuclease;
b. a recombinant site-directed nuclease operably linked to a supercharged
protein, wherein the site-directed nuclease comprises an RNA-binding portion
that interacts with the second nucleotide sequence of the gRNA and wherein
the site-directed nuclease specifically binds and cleaves the target DNA that
comprises the mutation associated with the T-cell disorder to create a double
stranded break in the target DNA; and
c. a single stranded donor oligonucleotide (ssODN) comprising a third
nucleotide sequence that hybridizes to a genomic sequence flanking the double
stranded break in the target DNA and that integrates into the target DNA to
correct the mutation associated with the T-cell disorder,
65. The complex of claim 64 wherein the supercharged protein is operably
linked to the
amino-terminus or the carboxy-terminus of the nuclease.
66. The complex of any of claims 64 or 65 wherein the recombinant site-
directed
nuclease operably linked to a supercharged protein further comprises a trans-
activating transcriptional activator (TAT) peptide operably linked to the
amino-
terminus of the site-directed nuclease.
67. The complex of any of claims 64-66, wherein the supercharged protein has
an overall
positive charge that is greater than its corresponding unmodified protein.
68. The complex of claim 67, wherein the overall positive charge is from about
+5 to
about about +40.
69. The complex of any of claims 64-68, wherein the supercharged protein is
superpositively charged green fluorescent protein (GFP).
70. The complex of claim 69, wherein the supercharged protein is a
superpositively
charged +36 GFP.
62

71. The complex of any of claims 64-70, wherein the nuclease is Cas9.
72. The complex of any of claims 64-71, wherein the molar ratio of gRNA to
site-directed
nuclease operably linked to a supercharged protein to ssODN is from about
1:1:0.2 to
about 1.5:1:2Ø
73. The complex of any of claims 1-15, wherein the recombinant site-directed
nuclease
operably linked to a supercharged protein is recombinant Cas9 operably linked
to
superpositively charged +36 GFP.
74. The complex of any of claims 1-15, wherein the recombinant site-directed
nuclease
operably linked to a supercharged protein is recombinant Cas9 operably linked
to
superpositively charged +36 GFP.
75. The complex of claim 74, wherein superpositively charged +36 GFP is
operably
linked to the carboxy-terminus of Cas9.
76. The complex of claim 74 or 75, wherein the ssODN hybridizes to the genomic
sequence encoding hemoglobin and comprises SEQ ID NO: 52.
63

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
CRISPR/CAS9 COMPLEX FOR GENOMIC EDITING
This application claims the benefit of U.S. Provisional Application No.
62/181,138,
filed June 17, 2015, and U.S. Provisional Application No. 62/266,316, filed
December 11,
2015, both of which are hereby incorporated herein in their entireties by this
reference.
BACKGROUND
Clustered regularly interspaced short palindromic repeats (CRISPR)-associated
(Cas)
systems (CRISPR-Cas9 systems) are used for gene editing at desired genomic
sites in
mammalian cells. In CRISPR-Cas9 systems, a Cas9 nuclease is targeted to a
genomic site by
complexing with a guide RNA that hybridizes to a target site in the genome.
This results in a
double-strand break that initiates either non-homologous end-joining (NHEJ) or
homology-
directed repair (HDR) of genomic DNA via a double-strand or single-strand DNA
repair
template. However, repair of a genomic site via HDR is inefficient.
SUMMARY
Provided herein is a complex for correcting a mutation in the genome of a cell
or
populations of cells. The complex comprises a guide RNA (gRNA) comprising a
first
nucleotide sequence that hybridizes to a target DNA in the genome of the cell,
wherein the
target DNA comprises a mutation, and a second nucleotide sequence that
interacts with a site-
directed nuclease. The complex further comprises a recombinant site-directed
nuclease
operably linked to a supercharged protein, wherein the site-directed nuclease
comprises an
RNA-binding portion that interacts with the second nucleotide sequence of the
guide RNA
and wherein the site-directed nuclease specifically binds and cleaves the
target DNA to create
a double stranded break. The complex also comprises a single-stranded donor
oligonucleotide (ssODN) that hybridizes to a genomic sequence flanking the
double stranded
break in the target DNA and integrates into the target DNA to correct a
mutation in the target
DNA.
Methods of site-specific modification of a target DNA in a cell or a
population of
cells are also provided. The methods comprise introducing a complex for
correcting a
mutation in the genome of the cell, wherein the complex is introduced into the
cells under
conditions that allow homology-directed repair (HDR) and integration of the
ssODN into the
target DNA. The method further provides for a high rate of cell survival in
corrected cells.
1

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
Further provided is a method of treating a disease associated with a mutation
in the
genomic sequence encoding hemoglobin in a subject. The method comprises
introducing
into a population of cells obtained from the subject a complex for correcting
a mutation in the
genomic sequence encoding hemoglobin under conditions that allow homology-
directed
repair (HDR) to correct the mutation in the genomic sequence encoding
hemoglobin and
transplanting the corrected cells into the subject.
DESCRIPTION OF THE FIGURES
Figures 1A-1C show that in vitro differentiation of JAK3 C1837T patient
induced
pluripotent stem cells (iPSCs) recapitulates SCID phenotypes. Figures 1A and
1B show flow
cytometry of iPSC-derived T cells. JAK3 WT iPSCs (Control) and JAK3- deficient
iPSCs
(JAK3 C1837T) were differentiated into CD34+ cells on 0P9 stromal cells and,
subsequently, into T cells on 0P9-DL4 monolayers. T-cell differentiation from
JAK3-
deficient iPSCs was absent compared to controls; no CD3+ T cells or CD3-
CD16+CD56+
NK cells were observed (Figure 1A), and no CD4+CD8+ double positive (DP), CD4+
single
positive (SP), or CD8+ single positive (SP) T cells were detected (Figure 1B).
Figure 1C
shows the results of RT-qPCR assays for transcripts of key genes that regulate
early events
during specification of the T cell lineage. RNA levels are shown relative to
GAPDH
expression.
Figures 2A-2C show that BCL2 partially rescues T cell developmental defects in
JAK3¨deficient, in-vitro derived cells. Figure 2A shows apoptosis of JAK3-
deficient, iPSC-
derived T cells compared to JAK3 WT controls. Annexin V-positive cells were
analyzed at T
cell induction day 10 (TD10) and 17 (TD17). Four independent experiments were
performed
with control JAK3 WT cells (Control) and 5 independent experiments were
performed with
JAK3-deficient cells (JAK3 C1837T). *P <0.005. Figure 2B shows the results of
RT-qPCR
assays for anti-apoptotic BCL2 and proapoptotic BAX expression in two lines (1
and 2) from
JAK3 WT (Control) and JAK3-deficient cells (JAK3 C1837T). ND, not determined
(due to
insignificant JAK3 qPCR signal). RNA levels are shown relative to GAPDH
expression.
Figure 2C shows flow cytometry of JAK3¨deficient iPSCderived T cells
transduced with
BCL2-2A-GFP lentivirus to assess effects on NK (CD16+56+) and T cell (CD3+)
development and DP (CD4+CD8+) to SP (CD4+ or CD8+) T cell maturation.
Figures 3A-3D show that CRISPR/Cas9 enhanced correction of the JAK3 C1837T
mutation in patient-specific iPSCs. Figure 3A depicts the strategy for genome
modification
2

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
using CRISPR/Cas9 to induce double-strand breaks in the JAK3 locus and a
template for
homology directed repair. Top line, structure of the JAK3 gene. Open boxes,
exons. Asterisk,
C1837T mutation. Arrows, guide RNAs. Figure 3B, top, shows PCR analysis
demonstrating
homologous recombination; primers for 5' and 3' analysis are indicated. (Lower
Left) RT-
PCR analysis demonstrating JAK3 mRNA expression in JAK3 WT (Control), JAK3-
deficient (JAK3 C1837T), and corrected (JAK3 Corrected) T cells. (Lower Right)
Western
Blot analysis demonstrating JAK3 protein expression in JAK3 WT (Control), JAK3-
deficient
(JAK3 C1837T), and corrected (JAK3 Corrected) T cells. Figure 3C provides a
summary of
targeting efficiencies of guide RNAs. (Figure 3D) Sanger sequencing of the PCR
amplicons
from parental JAK3 iPSCs (Left), heterozygous corrected (Middle) and
homozygous
corrected iPSCs (Right). The two heterozygous clones were corrected with gRNA2
+ wild
type Cas9, and the homozygous clone was corrected with gRNA1 + gRNA2 + nickase
Cas9
(D10A).
Figures 4A-4C show in vitro differentiation of JAK3 corrected patient iPSCs
produces T cells with phenotypic and functional characteristics of mature T
cells. Figure 4A
shows the expression of T cell developmental markers in JAK3 WT (Control,
n=3), JAK3-
deficient (JAK3 C1837T, n=5) and JAK3- corrected (JAK3 Corrected, n=6) T
cells. Cells
were stained with indicated antibodies and analyzed by flow cytometry at T
cell induction
Day 14, 21, 28 and 35 (TD 14, 21, 28 and 35). Figure 4B shows T cell receptor
(TCR)
analysis of JAK3-corrected T cells. A highly diverse repertoire of TCR Vfl is
represented in T
cells derived from corrected SCID patient iPSCs. Figure 4C shows flow
cytometry
demonstrating T cell activation in JAK3-corrected T cells. T cells derived
from JAK3 WT
(Control) and JAK3-corrected iPSCs were stimulated with anti-CD3/28 beads for
3 days
before analysis of activation markers CD25 and CD69. The data were gated on
CD3+
populations.
Figures 5A-5C show in vitro generation of CD34+ HSCs from hiPSCs by co-culture
with human bone marrow stromal cells (hMSC). Human iPSCs were cultured on
hMSCs for
18 days before analysis for hematopoietic markers, CD34 and CD43 (Figure A).
CD34+
cells were purified on beads and differentiated into T cells (Figure B),
erythroid and myeloid
cells (Figure C). To generate T cells, purified CD34+ cells were plated on 0P9-
DL4 cells for
3 to 4 weeks. For the CFC assay to generate myeloid and erythroid cells,
purified CD34+
cells were plated in MethoCult H4434 Classic medium according to the
manufacturer's
protocol. These data demonstrate that hiPSC can be efficiently differentiated
into multipotent
HSC after co-culture on hMSC.
3

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
Figure 6A-6C show in vitro generation of T cells by culturing hiPSC derived
CD34+
cells with hMSC-DL4. To generate CD7+ T progenitor cells, hiPSC derived CD34+
cells
were co-cultured on hMSC-DL4 for 3 to 4 weeks (Figure 6A). When CD7+ cells
from Figure
6A were purified on magnetic beads and co-cultured on 0P9-DL4, fully mature
CD4+/CD8+/CD3+/TCR-c43+ cells were produced in 10 days or less (Figures B and
C).
These data demonstrate that hiPSC can be efficiently differentiated into CD7+
lymphoid
progenitors after co-culture on hMSC-DL4.
Figure 7 shows in vitro generation of y6 T cells from hiPSC. Human iPSC were
transduced with a lentiviral vector carrying a pre-rearranged human V y61 cDNA
linked with
a 2A-GFP cDNA fragment. After co-culture with 0P9 for 18 days, hiPSC derived
CD34+
cells were purified on magnetic beads. These cells were subsequently plated on
0P9-DL4
cells for T cell differentiation. Cells were harvested at Day 32 and T cell
surface markers
were analyzed by FACS. The GFP+ population represents V61-2A-GFP lentiviral
transduced
cells. A high percentage of these GFP positive cells expressed V61 (66%). A
low percentage
of GFP negative cells expressed V61 (1%). These results demonstrate that V6 T
cells
expressing recombinant T Cell Receptors (TCR) can be efficiently produced from
genetically
modified iPSC. Production of V6 T cells expressing recombinant T Cell
Receptors (TCR)
specific for tumor antigens provides a powerful cellular therapy for many
types of cancer.
Figure 8 shows that a correction complex including guide RNA, a modified Cas9
and
a single stranded oligonucleotide donor sequence (ssODN) can correct a sickle
cell mutation.
The complex was introduced into sickle iPSC by nucleoporation, and 2 days
later genomic
DNA was analyzed by digital PCR (ddPCR) and sequenced. Over 65% of the cells
contained
at least one corrected gene. The results were confirmed as follows. Two days
after
introduction of the correction complex, the cells were plated in culture
dishes, and 43
individual iPSC colonies were isolated. Genomic DNA was isolated from these
colonies and
the beta-globin gene was sequenced. Sixty-five percent of the colonies
contained at least one
corrected beta-globin gene (S corrected to A).
Figure 9 shows that introduction of a sickle cell correction complex (gRNA-
modified
recombinant Cas9-ssODN) into patient primary bone marrow CD34+ cells can
correct a
sickle cell mutation. After twelve days of in vitro differentiation, DNA was
analyzed by
digital PCR (ddPCR) and sequenced. Approximately equal amounts of betaA and
betaS
mRNA were observed.
Figure 10 is an isoelectric focusing (IEF) gel of in vitro differentiated red
blood cells
from the corrected sickle patient CD34+ cells of Figure 9, showing an HbA
(normal
4

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
hemoglobin) to HbS (hemoglobin with sickle cell mutation) ratio of about 1:3,
which is
sufficient to inhibit sickling and treat sickle cell anemia.
Figure 11 shows that engineered positively charged Cas9 RNPs/ssODN (EpcCas9
RNPs/ssODN) efficiently correct the sickle mutation in human patient iPSCs.
Wild type
Cas9 (Cas9WT) RNP and eight engineered positively charged (EpcCas9) RNPs were
co-
nucleoporated with correction ssODN into human sickle iPSCs. Sickle correction
efficiencies in the pooled cells were determined by Sanger sequencing at two
days post
nucleofection. The arrow indicates the position of sickle correction (T->A)
and the scissors
indicate the Cas9WT-36GFP RNP cutting site on the sickle HBB DNA.
Figure 12 shows the results of deep sequencing of on-target modifications in
human
sickle iPSC populations. On-target deep sequencing analysis of human sickle
iPSCs
nucleoporated with Cas9WT RNP/ssODN, Cas9WT-EGFP, or four EpcCas9 RNPs/ssODNs
is shown. Black bars indicate the corrected base and the space below the black
bars indicates
the sickle cell mutation. The negative control and the ssODN alone both show
only the sickle
cell mutation. All iPSC samples also contain a SNP near the sickle mutation
(column on
right hand side).
Figure 13 shows that TAT-CAs9WT-EGFP RNP suppresses on-target indels. Human
sickle iPSCs were nucleoporated with Cas9WT and TAT-Cas9WT-EGFP RNPs with
(+ssODN) or without correction ssODN (-ssODN). Indel and correction
efficiencies were
analyzed by Sanger sequencing at two days post nucleoporation. The arrows
indicate the
position of sickle correction (T->A) and the scissors indicate the Cas9WT-
36GFP RNP
cutting site on the sickle HBB DNA.
Figure 14 shows that EpcCas9 RNPs suppress on-target indels in human sickle
iPSCs.
Human sickle iPSCs were nucleoporated with Cas9WT and five EpcCas9 RNPs, with
or
without correction ssODN. Indel and correction efficiencies were analyzed by
Sanger
sequencing at two days post nucleoporation. The arrows indicate the position
of sickle
correction (T->A) and the scissors indicate the Cas9WT-36GFP RNP cutting site
on the
sickle HBB DNA.
Figure 15 shows that EpcCas9 RNPs enhance cell survival after nucleoporation
in
human sickle iPSCs. Human sickle iPSCs were nucleoporated with Cas9WT RNP and
seven
EpcCas9 RNPs with or without correction ssODN. Cell survival was assessed by
light
microscopy at two days post nucleofection.
Figures 16A and 16B show ssODN:Cas9 RNP ratios for sickle correction in human
iPSCs. Correction ssODN and Cas9WT-36GFP/T2 RNP were nucleoporated into sickle
5

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
patient iPSC at molar ratios of 0, 0.2, 0.5, 1.0, 1.15, 1.35, 1.5 and 2Ø (A
Cas9WT-
36GFP:T2 gRNA molar ratio of 1:1.35 was fixed for these experiments. For
example, the
r=0.5 value in the graph below is 0.5 ssODN:1.0 Cas9WT-36GFP:1.35 T2 gRNA.)
Forty-
eight hours after nucleoporation of the ssODN:Cas9WT-36GFP RNPs, sickle
corrections
were quantitated by digital droplet PCR (ddPCR) (Fig. 16A) and Sanger
sequencing (Fig.
16B). The percent correction (betaA/betaS alleles x 100) was plotted verses r
(ssODN:Cas9WT-36GFP RNP). A dashed sigmoidal curve was fitted with the data
points.
(B) An arrow indicates the position of sickle correction (T->A) and scissors
indicate the
Cas9WT-36GFP RNP cutting site on sickle HBB DNA.
Figure 17 shows Cas9:sgRNA ratios for sickle correction in human iPSCs. Cas9-
36GFP:sgRNA molar ratios of 1:1.15, 1:1.35 and 1:1.50 with ssODN molar ratios
of 1.15 or
1.35 were tested to determine optimal correction efficiency of the sickle
mutation in patient
iPSC. The mixtures were nucleoporated into human sickle iPS cells and the
Sanger
sequencing results for the pooled cells were analyzed at two days post
nucleofection. Arrows
indicate the position of sickle correction (T->A) and scissors indicate the
Cas9WT-36GFP
RNP cutting site on sickle HBB DNA.
Figure 18 shows correction of human sickle iPSCs by EpcCas9 RNP/ssODN. Sanger
sequencing analysis of pooled human sickle iPS cells nucleofected with TAT-
Cas9WT-
36GFP-INF7 RNP/ ssODN was performed. The arrow indicates the position of
sickle
correction (T->A) and the scissors indicate the position of EpcCas9 RNP
induced DSB on the
sickle HBB DNA.
Figures 19A and 19B show correction of human iPSCs with EpcCas9 RNP and
wobble ssODNs. Human sickle iPSC were nucleoporated with TAT-Cas9-36GFP-INF7
RNP
and ssODNs containing wobble bases near the gRNA cleavage sites. (A) Sanger
sequencing
of iPSC populations nucleoporated with Ti gRNA and Ti-wb ssODN was performed.
The
arrow on the left hand side of Figure 19A indicates the position of sickle
mutation and the 3
arrows located downstream of the sickle mutation indicate positions of wobble
bases.
Scissors point to the Ti cleavage site. (B) Sanger sequencing of iPSC
populations
nucleoporated with T2 gRNA and T2-wb ssODN was performed. The arrow on the
left hand
side indicates the position of the sickle mutation and the 2 arrows downstream
of the sickle
mutation indicate the positions of wobble bases. Scissors point to the T2
cleavage site.
Figures 20A and 20B show the results of whole genome sequencing (WGS) analysis
of 4 iPSC clones corrected with TAT-Cas9WT-36GFP-INF7 RNP/ssODNs. (A) On-
target
6

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
sequence analysis demonstrates sickle correction and wobble-base substitution.
(B) WGS
off-target analysis of genomic loci with homology to Ti and T2 sgRNA is shown.
Figures 21A-D show gene correction of sickle patient bone marrow CD34+ HSPCs.
(A) Human sickle bone marrow CD34+ cells were nucleoporated by Cas9WT, Cas9WT-
36GFP and TAT-Cas9WT-3xTAT RNPs/ssODN. Gene correction efficiency for pooled
populations cells was analyzed six days after nucleofection. The arrow
indicates the position
of sickle correction (T->A) and the scissor indicate the Cas9WT-36GFP RNP
cutting site on
the sickle HBB DNA. (B) mRNA correction by RT-PCR and Sanger sequencing in
Cas9WT-36GFP nucleoporated sickle CD34+ cells that were harvested after 10-day
culturing
in erythroid differentiation media. (C) IEF Gel analysis of in vitro
differentiated RBCs from
Cas9WT-36GFP RNP/ssODN nucleofected sickle CD34+ cells. Human sickle child
patient
blood lysate (SS) and human normal adult blood lysate (AA) that represent HbF,
HbS and
HBA proteins were also loaded as controls. (D) Mass spectrometry analysis of
in vitro
differentiated RBCs derived from sickle CD34+ cells nucleofected with Cas9WT-
36GFP
RNP/ssODN. The peaks demonstrate signals from uncorrected HbS protein and
corrected
HbA protein.
Figures 22A-C show correction of colonies derived from single CD34+
progenitors.
(A) BFU-E and CFU-GEMM colonies derived from nucleoporated human sickle CD34+
cells. (B) Representative Sanger Sequencing results of colonies obtained from
human sickle
CD34+ cells after nucleoporation with TAT-Cas9WT-36GFP-INF7 RNP/ssODN. (C)
Colony survival after nucleoporation with Cas9WT, Cas9WT-36GFP, and TAT-Cas9WT-
3xTAT RNPs plus ssODNs.
Figure 23 is a graphical summary of deep sequencing data from Table 6.
Figure 24 shows non-specific modifications near Cas9WT RNP targeting site. BFU-
E
colonies from Cas9WT RNP/ssODN nucleoporated sickle CD34+ cells contain indels
that do
not appear to be initiated at the cutting site. The top sequence labeled
'Upstream' is
representative of non-specific modifications upstream of the expected cleavage
site. The
bottom sequence labeled 'Downstream' represents non-specific modifications
observed
downstream of the expected cleavage site. Arrows indicate the position of the
sickle
mutation and scissors indicate the expected cleavage site of Cas9WT RNP.
Figure 25 shows isoelectric focusing (IEF) gel analysis of blood six weeks
after
primary transplantation of sickle mouse fetal liver c-kit+ cells nucleoporated
with Cas9
RNP/ssODN into irradiated C57BI/6 mice to correct a sickle cell mutation.
Mouse fetal liver
c-kit+ cells are equivalent to human cord-blood Cd34+ cells.
7

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
Figure 26 shows ddPCR analysis of FACS purified bone marrow cells at twelve
weeks post-transplantation into irradiated C57BI6 mice. Twelve weeks after
nucleoporation
and transplantation, approximately 50% of erythroid cells (Ter119+) and
myeloid cells
(CD11b+ and CD11b+/GR1+) are corrected. Erythroid and myeloid cells are
relatively short
lived; therefore, these cells are derived from transplanted HSCs. Correction
levels in B and T
cells can rise to approximately 50% after secondary transplantation at twelve
weeks (twenty-
four weeks total). After twenty-four weeks, most if not all hematopoietic
cells will be
derived from long-term HSCs.
Figure 27 shows IEF gel analysis of blood from mice twelve weeks after primary
transplantation and six weeks after secondary transplantation of cells
nucleoporated with
Cas9 RNP/ssODN to correct a sickle cell mutation. Human HbA is produced in
mice after
transplantation of HSCs nucleoporated with Cas9 RNP/ssODN to correct a sickle
cell
mutation.
DETAILED DESCRIPTION
Provided herein are CRISPR/Cas9 complexes for genomic modification of cells.
Methods of using the complexes provided herein result in increased efficiency
of
modification, an increased cell survival ratio and/or an increased ratio of
HDR to NHEJ in the
cells. These complexes and methods can be used for therapeutic purposes, for
example, to
correct a mutation in cells, wherein the mutation is associated with a disease
or disorder.
Provided herein is a complex for correcting a mutation in the genome of a cell
comprising (a) a guide RNA (gRNA) comprising a first nucleotide sequence that
hybridizes
to a target DNA in the genome of a cell, wherein the target DNA comprises a
mutation, and a
second nucleotide sequence that interacts with a site-directed nuclease; (b) a
recombinant
site-directed nuclease operably linked to a supercharged protein, wherein the
site-directed
nuclease comprises an RNA-binding portion that interacts with the second
nucleotide
sequence of the guide RNA and wherein the site-directed nuclease specifically
binds and
cleaves the target DNA to create a double stranded break; and (c) a single-
stranded donor
oligonucleotide (ssODN) that hybridizes to a genomic sequence flanking the
double stranded
break in the target DNA and integrates into the target DNA to correct a
mutation in the target
DNA.
It is understood that the complex comprising a guide RNA (gRNA), a recombinant
site-directed nuclease and a donor nucleotide described herein does not occur
in nature. The
complex, however, provides the elements necessary with the required
configuration and
stoichiometry to efficiently and effectively modify cells. The gRNA molecule
binds to the
8

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
site-directed nuclease and targets the nuclease to a specific location within
the target DNA.
A gRNA comprises a first nucleotide sequence that hybridizes to a target DNA
in the genome
of a cell, wherein the target DNA comprises a mutation, and a second
nucleotide sequence
that interacts with a site-directed nuclease. The complexes described herein
can comprise
one or two separate gRNAs. Therefore, the term guide RNA includes both a
single guide
RNA and a double guide RNA. An example of a guide sequence that can be used to
correct a
mutation associated with sickle cell anemia is set forth herein as
TAACGGCAGACTTCTCCAC (SEQ ID NO: 1). An example of a guide sequence
comprising a stem loop for Cas9 binding is provided herein as
GTAACGGCAGACTTCTCCACGTTTTAGAGCTAGAAATAGCAAGTTAAAATAAGG
CTAGTCCGTTATCAACTTGAAAAAGTGGCACCGAGTCGGTGCTTTTTTT (SEQ ID
NO: 2). It is noted that the 5'G of SEQ ID NO: 2 was added by T7 during in
vitro
transcription.
In the complexes described herein, the recombinant site-directed nuclease can
be an
RNA-guided site-directed nuclease, for example, a Cas protein from any
bacterial species or
a functional fragment thereof For example, the Cas protein can be a Cas9
protein or a
functional fragment thereof As used herein, the term "Cas9" means a Cas9
protein or a
fragment thereof present in any bacterial species that encodes a Type II
CRISPR/Cas9
system. See, for example, Makarova et al. Nature Reviews, Microbiology, 9: 467-
477 (2011),
including supplemental information, hereby incorporated by reference in its
entirety. For
example, the Cas9 protein or a fragment thereof can be from Streptococcus
pyogenes. Full-
length Cas9 is an endonuclease that includes a recognition domain and two
nuclease domains
(HNH and RuvC, respectively). In the amino acid sequence, HNH is linearly
continuous,
whereas RuvC is separated into three regions, one left of the recognition
domain, and the
other two right of the recognition domain flanking the HNH domain. Cas9 from
Streptococcus pyogenes is targeted to a genomic site in a cell by interacting
with a guide
RNA that hybridizes to a 20-nucleotide DNA sequence that immediately precedes
an NGG
motif recognized by Cas9. This results in a double-strand break that is
repaired via HDR by a
donor nucleotide, for example, a ssODN or a double stranded DNA construct that
hybridizes
to a genomic sequence flanking the double stranded break in the target DNA and
integrates
into the target DNA to correct a mutation in the target DNA.
In the complexes provided herein, the molar ratio of gRNA to site-directed
nuclease
operably linked to a supercharged protein to ssODN can be from about 1:1:0.2
to about
1.5:1:2Ø For example, the molar ratio of gRNA to site-directed nuclease
operably linked to a
9

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
supercharged protein to ssODN can be about 1:1:1, 1.1:1:1, 1:1:1.15, 1:1:1.25,
1:1:1.30;
1:1:1.35; 1:1:1.40; 1:1:1.50, 1.2:1:1, 1.3:1:1. 1.4:1:1, 1.5:1:1, 1.5:1:1.15,
1.5:1:1.25,
1.5:1:1.35; 1.5:1:1.40, 1.5:1:1.45; 1.5:1:1.50; 1.5:1:1.55; 1.5:1:1.60;
1.5:1:1.65; 1.5:1:1.70;
1.5:1:1.75; 1.5:1:1.80; 1.5:1:1.85; 1.5:1:1.90; 1.5:1:1.95; 1.5:1: 2.0 or any
ratio in between
these ratios. Complexes having these molar ratios can be used in any of the
methods
described herein. Methods for preparing a complex prior to introducing the
complex into a
cell or a population of cells are set forth in the Examples.
As used herein, a supercharged protein can be a superpositively charged
protein that
has an overall positive charge that is greater than its corresponding
unmodified protein. For
example, the superpositively charged protein can be a superpositively charged
green
fluorescent protein (GFP) that has an overall positive charge from about +5 to
about +40. For
example, the overall positive charge can be about +5, +6, +7, +8, +9, +10,
+11, +12, +13,
+14, +15, +16, +17, +18, +19, +20, +21, +22, +23, +24, +25, +26, +27, +28,
+29, +30, +31,
+32, +33, +34, +35, +36, +37, +38, +39 or +40.
The supercharged protein can be operably linked to the amino-terminus or the
carboxy-terminus of the nuclease. It is also contemplated that the
supercharged protein can
be associated with the nuclease, without necessarily being covalently linked
to the nuclease.
An example of a supercharged protein is a superpositively charged GFP, for
example, +36
GFP. +36 GFP can be operably linked to the amino or carboxy- terminus of Cas9
or a
functional fragment thereof See, for example, McNaughton et al., "Mammalian
cell
penetration, siRNA transfection, and DNA transfection by supercharged
proteins," PNAS
106(15): 6111-6116. An example of a polypeptide comprising +36 GFP operably
linked to
the carboxy-terminus of Cas9 is provided herein as SEQ ID NO: 3.
The nuclease can also be operably linked to a supercharged protein and one or
more
positively charged peptides, for example, one or more transactivating
transcriptional activator
(TAT) peptide can be operably linked to the amino-terminus or the carboxy-
terminus of the
nuclease. For example,and not to be limiting, a superpositively charged
protein can be
operably linked to the carboxy-terminus of the nuclease and one or more TAT
peptides (for
example, 1X TAT, 2X TAT, 3X TAT, 4X TAT, etc.) can be operably linked to the
amino-
terminus of the nuclease. An example of polypeptide comprising a TAT peptide
operably
linked to the amino-terminus of the nuclease and a superpositively charged GFP
operably
linked to the carboxy-terminus of the nuclease is provided herein as SEQ ID
NO: 4.
Polypeptide sequences that are at least about 75% identical to SEQ ID NO: 3 or
SEQ ID NO:

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
4 are also provided. For example, polypeptide sequences that are at least
about 75%, 80%,
85%, 90%, 95%, 99% or any percentage in between are also provided.
The nuclease can also be operably linked to a supercharged protein and one or
more
negatively charged peptides, for example, a negatively charged peptide of
about 10 to about
25 amino acids in length, for example, SEQ ID NO: 50, can be operably linked
to the
carboxy-terminus of the site-directed nuclease. For example,and not to be
limiting, a
superpositively charged protein can be operably linked to the carboxy-terminus
of the
nuclease and a negatively charged peptide can be operably linked to the
carboxy-terminus of
the superpositively charged protein.
As used throughout, recombination is a process of exchange of genetic
information
between two polynucleotides. Homology-directed repair (HDR) refers to DNA
repair that
takes place, for example, during repair of double-strand breaks in cells. This
process requires
nucleotide sequence homology and uses a donor molecule, for example, a single
stranded or a
double stranded nucleotide sequence as a template for repair of a target
genomic sequence,
i.e., the genomic sequence with the double-strand break, and leads to the
transfer of genetic
information from the donor to the target genomic sequence. Homology-directed
repair can
result in a modification of the sequence of the target genomic sequence. For
example, HDR
can result in an insertion, a deletion or a mutation in the target genomic
sequence. Part or all
of the sequence of the donor polynucleotide can be incorporated into the
target DNA. It is
also contemplated that the donor polynucleotide, a portion of the donor
polynucleotide, a
copy of the donor polynucleotide, or a portion of a copy of the donor
polynucleotide
integrates into the target DNA.
As used throughout, by non-homologous end joining (NHEJ) is meant the repair
of
double-strand breaks in DNA by direct ligation of the break ends to one
another without the
need for a homologous template (in contrast to homology-directed repair, which
requires a
homologous sequence to guide repair).
The complexes and methods provided herein can be used to correct any mutation
in a
target DNA by HDR. For example, and not to be limiting, the complexes can be
used to
replace an incorrect nucleotide sequence with a correct nucleotide sequence
(e.g., to restore
function to a target polynucleotide sequence that is impaired due to a loss of
function
mutation, i.e., a SNP) at a specific site in the genome. These mutations can
be associated
with an autoimmune disorder, a genetic disease, a blood disorder, a T cell
disorder, a
monogenic disorder, cancer, a neurodegenerative disease, a cardiovascular
disease or an
infectious disease, to name a few. For example, and not to be limiting, the
complexes and
11

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
methods provided herein can be used to correct a mutation associated with
sickle cell disease
(i.e., a mutation in a hemoglobin gene, for example, a GAG to GTG mutation at
codon 6 of
the beta-globin gene that results in a glutamic acid to valine substitution),
severe combined
immunodeficiency (SCID) (for example, a mutation in JAK3), beta thalassemia or
Wiskott-
Aldrich Syndrome.
Correction of single mutations or multiple mutations can be performed with one
or
more complexes. The complexes and methods provided herein can also be used to
insert
sequences into a specific site in the genome to correct a deletion, as opposed
to making a
correction or a substitution. The complexes and methods provided herein can
also be used to
insert a nucleotide sequence that encodes an a functional polypeptide into a
specific site in
the genome of the cell, in order to express the functional polypeptide in the
cell. The
functional polypeptide can be a polypeptide that is endogenous (i.e., normally
expressed by
the cell) or exogenous to the cell (i.e. not normally expressed by the cell).
For example,
chimeric antigen receptor (CAR) sequences can be inserted into the genome of a
T cell
precursor in order to generate cancer specific T cells for the treatment of
cancer. In another
example, the complexes and methods provided herein can be used to inhibit the
activity of a
gene at a specific site in the genome of the cell. For example, the complexes
and methods
provided herein can be used to insert sequences into the CXCR4 or CCR5
receptor to treat or
prevent HIV infection.
The complexes provided herein can modify or alter target DNA with surprisingly
high
efficiency as compared to conventional CRISPR/Cas systems. The efficiency of
alteration in
a population of cells can be at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%,
40%, 45%,
50%, 55%, 60%, 65%, 70%, 75% or 80% or higher or any percentage in between
these
percentages. The efficiency of alteration can also be greater than or equal to
about 80%.
Therefore, also provided herein are populations of cells, wherein at least 5%,
10%, 15%,
20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75% or 80% or higher or
any
percentage in between are altered. For example, a mutation associated with
sickle cell
disease or another disorder has been corrected. If a population of cells
comprising a mutation
associated with sickle cell disease is contacted with a CRISPR/Cas complex
described herein
and the mutation is corrected in about 5% of the cells, the efficiency of
modification or
alteration is about 5%. Optionally, a population of cells wherein the mutation
associated with
sickle cell disease is corrected in about 30% of the cells, including, for
example, 27%, 28%
and 29% is sufficient to treat sickle cell disease, upon transplantation in a
subject with sickle
cell disease. Optionally, a mutation associated with sickle cell disease is
corrected in about
12

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
40%, 50%, 60%, 70%, 80%, 90% or higher or any percentage in between, of the
cells in the
population.
In addition to altering the target DNA with high efficiency, the complexes
provided
herein can also increase the ratio of HDR to NHEJ in a population of cells
contacted with the
complex. The HDR/NHEJ ratio can be from about 10 to about 0.5. For example,
the
HDR/NHEJ ratio can be about 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, 0.5 or less or any
ratio in between
these ratios. In addition to high efficiency of correction and high rate of
HDR to NHEJ, the
cell survival rate for corrected cells can be at least about 50%, 60%, 70%,
80%, 90% or
higher and any percentage in between.
Any cell(s) can be modified or derived using the complexes described herein.
Introduction of the complex into the cells can be cell cycle dependent or cell
cycle
independent. Methods of synchronizing cells to increase the proportion of
cells in a particular
phase, for example, the S-phase , are known in the art. See, for example,
Takahashi et al.
"Efficient introduction of a gene into hematopoietic cells in S-phase by
electroporation," Exp.
Hematol. 19(5):343-346 (1991). Depending on the type of cell to be modified,
one of skill in
the art can readily determine if cell cycle synchronization is necessary.
The cell(s) can be a eukaryotic cell, for example, a mammalian cell. The cell
can also
be prokaryotic or a plant cell. The cell can be a human cell. The cell can be
a germ cell, a
somatic cell, a stem cell, a precursor cell or a progenitor cell. The
precursor cell can be, for
example, a pluripotent stem cell or a multipotent stem cell, like a
hematopoietic stem cell. As
used throughout, pluripotent cells include induced pluripotent stem cells.
Methods of making
induced pluripotent stem cells and known in the art and described in the
Examples. The cell
can also be CD34+ cell, optionally derived from an induced pluripotent stem
cell. The
CD34+ cell can be selected from the group consisting of a primary CD34+
hematopoietic
progenitor cell, a CD34+ peripheral blood cell, a CD34+ cord blood cell and a
CD34+ bone
marrow cell. The cell can also be a primary cell, for example, a primary CD34+
hematopoietic progenitor cell. The cells are cells that are not cancer cells,
cells that are not
tumor cells or cells that are not transformed cells. Cells can be screened
before or after
correction for evidence of undesirable genetic characteristics. Further
provided is a cell
comprising any of the complexes described herein. The cell can be in vitro, ex
vivo or in
vivo.
Further provided is a method of site-specific modification of a target DNA in
a
population of cells comprising introducing into the cells any of the complexes
described
herein, wherein the complex is introduced into the cells under conditions that
allow
13

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
homology-directed repair (HDR) and integration of a donor nucleotide, for
example, a
ssODN or double stranded nucleotide sequence into the target DNA. The complex
can be
introduced into the cell via nucleoporation. Methods for nucleoporation are
known in the art.
See, for example, Maasho et al. "Efficient gene transfer into the human
natural killer cell line,
NKL, using the amaxa nucleofection system," Journal of Immunological Methods
284(1-2):
133-140 (2004); and Aluigi et al. "Nucleofection is an efficient non-viral
transduction
technique for human bone marrow derived mesenchymal stem cells," Stem Cells
24(2): 454-
461 (2006)), both of which are incorporated herein in their entireties by this
reference.
In some of the methods provided herein, the donor nucleotide, for example, a
ssODN
or a double stranded nucleotide sequence integrates into a target DNA and
corrects a
mutation in the target DNA. In the methods provided herein the ratio of HDR to
NHEJ in a
population of cells is increased relative to other CRISPR-Cas9 delivery
methods. The
HDR/NHEJ ratio can be from about 10 to about 0.5. For example, the HDR/NHEJ
ratio can
be about 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, 0.5 or less or any ratio in between
these ratios. In the
methods provided herein, the efficiency of alteration by HDR can be at least
about 5%, 10%,
15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80% or
greater or
any percentage in between these percentages. The efficiency of alteration by
HDR can also
be greater than or equal to about 80%. For example, if a population of cells
comprising a
mutation associated with sickle cell anemia is contacted with a CRISPR/Cas
complex
described herein and the mutation is corrected in about 5% of the cells, the
efficiency of
alteration by HDR is about 5%. The population of cells can be obtained from
the subject
having a disorder such that at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%,
40%, 45%,
50%, 55%, 60%, 65%, 70%, 75% or 80% or greater or any percentage in between
these
percentages, of the cells undergo HDR to correct a mutation associated with
the disorder. In
some cases greater than 80% of the cells from the subject will undergo HDR to
correct a
mutation associated with the disorder. In the methods described herein,
between about 50%
and 99% of the cells survive after introduction of the complex. For example,
great than about
50%, 60%, 70%, 80%, 90%, 95%, 99% or any percentage in between these
percentages, of
corrected cells survive after introduction of the complex.
Further provided is a method of treating a disease associated with a mutation
in the
genomic sequence encoding hemoglobin in a subject comprising: (a) introducing
into a
population of cells obtained from the subject a complex comprising (1) a guide
RNA (gRNA)
comprising a first nucleotide sequence that hybridizes to a target DNA in the
genome of a
cell, wherein the target DNA is a hemoglobin gene that comprises a mutation,
and a second
14

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
nucleotide sequence that interacts with a site-directed nuclease; (2) a
recombinant site-
directed nuclease operably linked to a supercharged protein, wherein the site-
directed
nuclease comprises an RNA-binding portion that interacts with the second
nucleotide
sequence of the guide RNA and wherein the site-directed nuclease specifically
binds and
cleaves the target DNA to create a double stranded break; and (3) a single-
stranded donor
oligonucleotide (ssODN) that hybridizes to a genomic sequence flanking the
double stranded
break in the target DNA and integrates into the target DNA to correct the
mutation in
hemoglobin gene; and (b) transplanting the corrected cells into the subject.
In the methods for treating a disease associated with a mutation in the
genomic
sequence encoding hemoglobin in a subject, for example, sickle cell anemia,
the subject with
sickle cell anemia can optionally be a transfusion dependent subject or a
subject with at least
one silent infarction. The subject can also be less than about twelve months,
eleven months,
ten months, nine months, eight months, seven months, six months, five months,
four months,
three months, two months, or one month in age. As infants are routinely screen
for sickle cell
disease, infants can be treated before symptoms of the disease manifest. The
methods
provided herein can further comprise diagnosing a subject with a disorder, for
example,
sickle cell disease.
As set forth above, cells can be obtained from the subject with the disease or
from a
related donor. For example, bone marrow cells can be obtained or harvested
from the subject
Bone marrow harvesting involves collecting stem cells with a needle placed
into the soft
center of the bone, the marrow. Bone marrow can be harvested for example, from
the hip
bones or sternum of the subject. From about 500 ml to about 1 liter of bone
marrow can be
obtained from the subject.
In any of the methods provided herein the cell(s) can be a eukaryotic cell,
for
example, a human cell. The cell can be a germ cell, a stem cell, a precursor
cell. The
precursor cell can be, for example, a pluripotent stem cell or a hematopoietic
stem cell. As
used throughout, pluripotent cells include induced pluripotent stem cells.
Methods of making
induced pluripotent stem cells and known in the art and described in the
Examples. The cell
can also be CD34+ cell. The CD34+ cell can be selected from the group
consisting of a
primary CD34+ hematopoietic progenitor cell, a CD34+ peripheral blood cell, a
CD34+ cord
blood cell and a CD34+ bone marrow cell. The cell can also be a primary cell,
for example, a
primary CD34+ hematopoietic progenitor cell. The cells are that are not cancer
cells, cells
that are not tumor cells or cells that are not transformed cells. The cell can
be in vitro or ex
vivo. The cells can also be in a pharmaceutically acceptable composition.

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
The methods provided herein can further comprise culturing the cells corrected
with
HDR. For example, the cells can be cultured under conditions for expansion or
under
conditions that promote differentiation of the corrected cells into T-cells.
For example, and
not to be limiting, using the methods provided herein, after a mutation has
been corrected in
induced pluripotent stem cells via HDR, the corrected cells can be co-cultured
with human
bone marrow stromal cells to generate CD34+ cells. The CD34+ cells can then be
cultured
under conditions that differentiate the CD34+ cells into T cells.
The methods provided herein can further comprise screening the corrected cells
for
the proper correction, other mutations, or NEJ prior to transplantation.
Optionally cells can
be screened to detect cells with one or more corrections.
In the methods provided herein, the cells can be transplanted into the subject
after
modification, for example, after correction of a mutation by HDR. The cells
can be
transplanted into the subject with or without differentiation. For example,
modified
hematopoietic stem cells (HSCs) can be administered in a bone marrow
transplant, wherein
the HSCs are allowed to differentiate and mature in vivo in a subject
Alternatively, the
modified cells can be differentiated into a desired population of cells prior
to transplantation.
As used herein, transplanting, introducing or administering cells to a subject
refers to
the placement of cells into a subject. For example, the cells described herein
comprising a
target DNA sequence corrected or modified according to the methods described
herein can be
transplanted into a subject, by an appropriate route which results in at least
partial
localization of the transplanted cells at a desired site. The cells can be
implanted directly to
the desired site, or alternatively can be administered by any appropriate
route which results in
delivery to a desired location in the subject where at least a portion of the
implanted cells
remain viable. For example, the cells can be administered systemically, via
intravenous
infusion. The period of viability of the cells after administration to a
subject can be as short
as a few hours, e. g. twenty-four hours, to a few days, to as long as several
years.
For ex vivo methods, cells can be autologous cells, i.e., a cell or cells
taken from a
subject who is in need of modification of a target DNA in the cell or cells
(i.e., the donor and
recipient are the same individual). As described herein, the modification can
be, for example
correction of a mutation, insertion of a sequence that inhibits activity of a
protein or insertion
of a sequence that increases expression of a protein, for example, insertion
of a sequence
encoding a chimeric antigen receptor that can be used to target cancer cells.
Autologous cells
can be used to avoid immunological reactions that can result in rejection of
the cells. In other
words, when using autologous cells, the donor and recipient are the same
subject.
16

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
Alternatively, the cells can be heterologous, e.g., taken from a donor,
preferably a related
donor. The second subject can be of the same or different species. Typically,
when the cells
come from a donor, they will be from a donor who is sufficiently
immunologically
compatible with the recipient to reduce the chances of transplant rejection,
and/or to reduce
the need for immunosuppressive therapy. The cells can also be obtained from a
xenogeneic
source, i.e., a non-human mammal that has been genetically engineered to be
sufficiently
immunologically compatible with the recipient, or the recipient's species. Any
of the
methods of treating a disorder described herein can further comprise
administering one or
more immunosuppressants to the subject.
In the methods involving transplantation, a subject optionally undergoes
myeloablative therapy prior to transplantation of any of the cells described
herein. The
myeloablative therapy can include administering one or more doses of
chemotherapy,
radiation therapy, or both, that results in severe or complete depletion of
healthy bone
marrow cells. In another example, the subject can undergo submyeloablative
therapy that
includes administering one or more doses of chemotherapy, radiation therapy,
or both, that
depletes a portion of the healthy bone marrow cells. The cells can also be
transplanted into
subjects that have undergone nonablative chemotherapy. For example, the cells
can be
transplanted into a subject that has been treated with Busulfan, Fludarabine
and/or
Treosulfan.
In the methods involving transplantation, an effective dose or amount of
corrected
cells is administered to the subject. The terms effective amount and effective
dosage are used
interchangeably. The term effective amount is defined as any amount necessary
to produce a
desired physiologic response. In some methods, about 1 X 106 to about 7 X 106
corrected
cells/kg can be administered, but this amount can vary depending on the
associated disorder.
The percentage of corrected cells that Effective amounts and schedules for
administering the
cells may be determined empirically, and making such determinations is within
the skill in
the art. The dosage ranges for administration are those large enough to
produce the desired
effect (e.g., treatment of a disease, for example, sickle cell anemia). The
dosage should not
be so large as to cause substantial adverse side effects, such as unwanted
cross-reactions,
anaphylactic reactions, and the like. Generally, the dosage will vary with the
age, condition,
sex, type of disease, the extent of the disease or disorder, route of
administration, or whether
other drugs are included in the regimen, and can be determined by one of skill
in the art. The
dosage can be adjusted by the individual physician in the event of any
contraindications.
17

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
Dosages can vary, and the agent can be administered in one or more dose
administrations
daily, for one or multiple days as needed.
As used throughout, a subject can be a vertebrate, more specifically a mammal
(e.g., a
human, horse, cat, dog, cow, pig, sheep, goat, mouse, rabbit, rat, and guinea
pig). The term
does not denote a particular age or sex. Thus, adult and newborn subjects,
whether male or
female, are intended to be covered. As used herein, patient or subject may be
used
interchangeably and can refer to a subject with or at risk of developing a
disorder. The term
patient or subject includes human and veterinary subjects.
As used herein the terms treatment, treat, or treating refers to a method of
reducing
one or more of the effects of the disorder or one or more symptoms of the
disorder, for
example, sickle cell disease, by eliciting an immune response in the subject.
Thus in the
disclosed method, treatment can refer to a 10%, 20%, 30%, 40%, 50%, 60%, 70%,
80%,
90%, or 100% reduction in the severity of sickle cell disease and other
disorders. For
example, a method for treating sickle cell disease is considered to be a
treatment if there is a
10% reduction in one or more symptoms of the infection in a subject as
compared to a
control. Thus the reduction can be a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%,
90%,
100%, or any percent reduction in between 10% and 100% as compared to native
or control
levels. It is understood that treatment does not necessarily refer to a cure
or complete
ablation of the disorder or symptoms of the disorder.
Also provided is a method of correcting a mutation associated with a T-cell
disorder
comprising introducing into a population of cells obtained from a subject with
the T-cell
disorder a complex comprising: (a) a guide RNA (gRNA) comprising a first
nucleotide
sequence that hybridizes to a target DNA in the genome of a cell, wherein the
target DNA
comprises the mutation associated with the T-cell disorder, and a second
nucleotide sequence
that interacts with a site-directed nuclease; (b) a recombinant site-directed
nuclease operably
linked to a supercharged protein, wherein the site-directed nuclease comprises
an RNA-
binding portion that interacts with the second nucleotide sequence of the gRNA
and wherein
the site-directed nuclease specifically binds and cleaves the target DNA that
comprises the
mutation associated with the T-cell disorder to create a double stranded break
in the target
DNA; and (c) a single stranded donor oligonucleotide (ssODN) comprising a
third nucleotide
sequence that hybridizes to a genomic sequence flanking the double stranded
break in the
target DNA and that integrates into the target DNA to correct the mutation
associated with
the T-cell disorder, wherein the complex is introduced into the cell under
conditions that
18

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
allow homology-directed repair (HDR) to correct the mutation associated with
the T-cell
disorder.
In the methods provided herein, the target DNA comprising a mutation
associated
with a T-cell disorder can be a target DNA that encodes a protein associated
with T-
lymphocyte development. For example, the target DNA can encode JAK3. Such
corrected
cells can be used, for example, in the treatment of SCID.
In addition to correcting mutations in the genome of a cell, the complexes and
methods provided herein can also be used to insert functional polypeptides at
specific sites in
the genome of a cell, such that the polypeptide is expressed by the cell. The
polypeptide can
be expressed in the cell or on the cell surface.
Also provided is a method of making tumor-specific T-cell precursor cells
comprising
introducing into a population of T-cell precursor cells a complex comprising:
(a) a guide
(gRNA) comprising a first nucleotide sequence that hybridizes to a target DNA
in the
genome of the T cell precursor cells and a second nucleotide sequence that
interacts with a
site-directed nuclease; (b) a recombinant site-directed nuclease operably
linked to a
supercharged protein, wherein the site-directed nuclease comprises an RNA-
binding portion
that interacts with the second nucleotide sequence of the gRNA and wherein the
site-directed
nuclease specifically binds and cleaves the target DNA to create a double
stranded break; and
(c) donor nucleotide sequence comprising a third nucleotide sequence that
encodes a chimeric
antigen receptor (CAR) and a fourth nucleotide sequence that hybridizes to a
genomic
sequence flanking the double stranded break in the target DNA, wherein the
complex is
introduced into the T-cell precursor cells under conditions that allow
homology-directed
repair (HDR) and integration of the third nucleotide sequence into the target
DNA to form
modified T-cell precursor cells that express the CAR.
The T cell precursor cells can be obtained from a subject with cancer. As set
forth
above, the HDR/NHEJ ratio can be from about 10 to about 0.5. For example, the
HDR/NHEJ
ratio can be about 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, 0.5 or any ratio in between
these ratios. In the
methods provided herein, the efficiency of alteration by HDR can be at least
about 5%, 10%,
15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80% or any
percentage in between these percentages. The efficiency of alteration by HDR
can also be
greater than or equal to about 80%. For example, when using the methods
described herein, if
a nucleotide sequence encoding an functional polypeptide, for example, a
nucleotide
sequence that encodes a CAR, is inserted in about 5% of the cells, the
efficiency of alteration
by HDR is about 5%. The population of cells can be obtained from the subject
that has
19

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
cancer such that at least about 5%, 10%, 15%, 20%, 25%, 30%, 350, 40%, 450,
50%, 550
,
60%, 65%, 70%, 7500 or 80% or any percentage in between these percentages, of
the cells
undergo HDR to insert a nucleotide sequence that encodes a chimeric antigen
receptor (CAR)
and form cells that express the CAR. In some cases greater than 80% of the
cells from the
subject will undergo HDR to correct a mutation associated with the disorder.
The modified T-cell precursor cells that express the CAR can be transplanted
into a
subject with cancer. As used herein, cancer is a disease characterized by the
rapid and
uncontrolled growth of aberrant cells. Cancer cells can spread locally or
through the
bloodstream and lymphatic system to other parts of the body. Examples of
cancers include
but are not limited to, breast cancer, prostate cancer, ovarian cancer,
cervical cancer, skin
cancer, pancreatic cancer, colorectal cancer, renal cancer, liver cancer,
brain cancer,
lymphoma, leukemia, lung cancer and the like. The modified T-cell precursor
cells that
express the CAR exhibit anti-tumor immunity when the antigen binding domain
binds to its
corresponding antigen.
Disclosed are materials, compositions, and components that can be used for,
can be
used in conjunction with, can be used in preparation for, or are products of
the disclosed
methods and compositions. These and other materials are disclosed herein, and
it is
understood that when combinations, subsets, interactions, groups, etc. of
these materials are
disclosed that while specific reference of each various individual and
collective combinations
and permutations of these compounds may not be explicitly disclosed, each is
specifically
contemplated and described herein. For example, if a method is disclosed and
discussed and
a number of modifications that can be made to a number of molecules including
the method
are discussed, each and every combination and permutation of the method, and
the
modifications that are possible are specifically contemplated unless
specifically indicated to
the contrary. Likewise, any subset or combination of these is also
specifically contemplated
and disclosed. This concept applies to all aspects of this disclosure
including, but not limited
to, steps in methods using the disclosed compositions. Thus, if there are a
variety of
additional steps that can be performed, it is understood that each of these
additional steps can
be performed with any specific method steps or combination of method steps of
the disclosed
methods, and that each such combination or subset of combinations is
specifically
contemplated and should be considered disclosed.
Publications cited herein and the material for which they are cited are hereby
specifically incorporated by reference in their entireties.

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
EXAMPLES
Example 1
Correction of SCID by CRISPR/Cas9 enhanced gene replacement
Mutations of the Janus family kinase JAK3 gene cause severe combined
immunodeficiency (SCID). JAK3 deficiency in humans is characterized by the
absence of
circulating T cells and natural killer (NK) cells with normal numbers of
poorly functioning B
cells (T¨B+NK¨). As shown herein, using SCID patient-specific induced
pluripotent stem
cells (iPSCs) and a T cell in vitro differentiation system, a complete block
in early T cell
development of JAK3-deficient cells was demonstrated. Correction of the novel
JAK3
mutation by CRISPR/Cas9 enhanced gene replacement restores normal T cell
development,
including the production of mature T-cell populations with a broad T Cell
Receptor (TCR)
repertoire. Whole genome sequencing of corrected cells demonstrated no
CRISPR/Cas9 off-
target modifications. Thus, provided herein is a novel approach for the study
of human
lymphopoiesis and a method for gene replacement therapy in humans with
immunodeficiencies.
Allogeneic hematopoietic stem cell (HSC) transplantation is currently the only
established therapy for SCID; however, delayed immune recovery and risk of
graft-vs-host
disease present significant risks. Treatment by retroviral-based gene therapy
has been
successfully demonstrated for X-linked SCID. However, severe adverse effects
of insertional
mutagenesis have been observed with retroviral gene therapy. Self-inactivating
lentiviral
vectors have been used effectively in recent clinical trials, but long-term
follow-up is needed
to thoroughly address safety concerns.
Provided herein is an alternative therapeutic strategy in which patient-
specific induced
pluripotent stem cells (iPSCs) are derived, and disease-causing mutations are
corrected by
gene replacement using a CRISPR-Cas9 complex. These corrected iPSCs could
optionally
be differentiated into hematopoietic progenitors for transplantation into
patients to treat the
disease (Hanna et al., "Treatment of sickle cell anemia mouse model with iPS
cells generated
from autologous skin," Science 318: 1920-1923 (2007)). As shown herein,
differentiation of
JAK3-deficient human T cells is blocked at an early developmental stage. Also
demonstrated
is that correction of the human JAK3 mutation by CRISPR/Cas9 enhanced gene
replacement
restores the differentiation potential of early T cell progenitors. These
corrected progenitors
are capable of producing NK cells and mature T cell populations expressing a
broad
21

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
repertoire of T-cell antigen receptors (TCR). These studies establish a
powerful system for
determining the mechanism of immunodeficiency in human SCID patients and for
testing
pharmacological and genetic therapies for the disorder.
Patient Information
The male patient was enrolled in an Institutional Review Board-approved study
in
accordance with the Declaration of Helsinki. The family history was negative
for immune
deficiencies. For the first 8 months of age he had poor weight gain, diarrhea,
and recurrent
bronchiolitis requiring frequent hospitalization. He was admitted to the
hospital at 8 months
of age with severe respiratory distress and oral thrush. Bronchoscopy with
bronchial alveolar
lavage demonstrated bacterial (pseudomonas, H flu, S. pneumonia) and viral
organisms
(respiratory syncytial virus). Immunologic evaluations demonstrated severe
hypogammaglobulinemia, with an IgE<3, IgA<4, IgG=29, IgM=26. Immune
phenotyping of
peripheral blood demonstrated complete absence of CD3+ T cells and NK cells,
though B
cells were present (absolute B cell count=875). Mitogen studies demonstrated a
complete
lack of response to concanavalin A, poke weed mitogen and phytohemagglutinin
A. The
diagnosis of SCID was confirmed by genetic testing, with a homozygous C>T
nucleotide
substitution in exon 14 of the JAK3 gene, resulting in the replacement of an
arginine codon
(CGA) with a stop codon (TGA) at amino acid position 613. This is the first
report linking
this JAK3 variant (rs149316157) to a clinical case of SCID. The patient
underwent a reduced
intensity conditioning matched unrelated bone marrow transplant, and is doing
well now two
years off therapy with complete immune reconstitution.
Human iPSC reprogramming and characterization
For iPSC induction, 5x104 primary keratinocytes were seeded into one well of a
6-
well plate. On the following day, keratinocytes were transduced with 1 mL of
virus
supernatant and 1 mL of human keratinocyte medium containing polybrene at a
final
concentration of 4 1.tg/mL. The keratinocytes were spinfected at 800 x g for
45 minutes (day
1). The transduction procedure was repeated again the next day. On day 3,
cells were changed
to fresh human keratinocyte medium and cultured for two more days. On day 5,
the
keratinocytes were trypsinized and transferred to a 10 cm dish pre-seeded with
mitomycin C-
treated murine embryonic fibroblasts (EFs) and cultured in human keratinocyte
medium.
On day 7, cells were changed to human ES medium and continuously cultured in
the same
dish for 3-4 weeks. ES medium was changed daily. Potential iPSC colonies were
visible after
2-3 weeks. These colonies were individually picked and expanded on MEFs for
analysis. To
22

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
remove the integrated lentiviral and polycistronic sequences, iPSCs were
infected with a Cre-
expressing adenovirus (rAd-Cre-IE). Individual colonies were picked and Cre-
mediated
removal of foxed sequences was verified by PCR using the primers
gctaattcactcccaaagaagacaag (SEQ ID NO: 5) and cttcagcaagccgagtcctg (SEQ ID NO:
6).
Generation of CD34+ cells and T cells with 0P9 co-culture
The procedure was described previously (Chang et al., "Broad T-cell receptor
repertoire in T-lymphocytes derived from human induced pluripotent stem
cells," PloS one 9,
e97335 (2014)). This method was used with the following modifications.
Cultures of hiPSCs
in one well of a 6 well plate were treated as described by Ohnuki et al
(Ohnuki M,
"Generation and characterization of human induced pluripotent stem cells. Curr
Protoc Stem
Cell Blot Chapter 4: Unit 4A 2 (2009)) with CTK solution to make small cell
clumps. Cell
clumps were then transferred to a 10 cm plate that was pre-seeded with 2-day
old 0P9 cells
in a-MEM-based medium containing 10% FBS, 1X penicillin/streptomycin and 100
[NI
mono-thioglycerol. The medium was changed every other day, and cells were
cultured for 18
days without splitting. After 18 days of co-culture, cells were harvested by
treating with
dissociation solution (0.15% collagenase IV and 0.015% hyaluronidase in a-MEM
medium)
for about 30 minutes and followed by 0.25% trypsin for another 30 minutes.
CD34+ cells
were then purified on anti- CD34+ magnetic beads (MicroBead Kit; Miltenyi
Biotec,
Bergisch Gladbach, Germany). For T cell differentiation, these CD34+ cells
were plated onto
0P9-DL4 cells and cultured with a-MEM medium containing 20% FBS, 5 ng/mL hFlt3-
L, 5
ng/mL hIL-7, and 10 ng/mL hSCF. The medium was changed every other day, and
cells were
transferred to new 0P9-DL4 plates every 4 days.
T cell stimulation
In vitro derived T cells from hiPSCs were stimulated by incubation with CD3/28
beads (Invitrogen, Carlsbad, CA) according to the manufacturers' protocol for
3 days prior to
analysis by flow cytometry, as previously described (Chang et al., 2014).
Flow Cytometry
Cells were harvested and washed before analysis with an LSRFortessa cell
analyzer
(BD Bioscience, San Jose, CA). For cell surface staining, propidium iodide
(PI, Sigma-
Aldrich, St. Louis, MO) was used to exclude dead cells. For apoptosis assay,
harvested cells
were first stained with cell surface antibodies for 30 min. After washing once
with 1X PBS,
the cells were resuspended in 100 [EL of Annexin Binding Buffer (Invitrogen,
Carlsbad, CA)
containing Annexin V-647 (Invitrogen, Carlsbad, CA) and PI and incubated for
15 min
before adding 400 [EL of Annexin Binding Buffer with PI. Antibodies were
obtained from BD
23

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
Biosciences unless otherwise indicated: CD3 (Percp-Cy5-5, clone UCHT1), CD4
(PE-Cy7,
clone SK3), CD7 (APC, BV510, clone M-T701), CD8 (APC-Cy7, clone SK1), CD16
(PE,
clone B73.1), CD25 (FITC, clone 2A3), CD34 (PE-Cy7, clone WM59), CD43 (PE,
clone
1G10), CD56- PE (clone MY31), CD69 (FITC, clone L78), NKG2D-PE (clone 1D11),
TCR-
c43 (FITC, PE, clone T10B9.1A-31), TCR-V61-FITC (Fisher Scientific,
Pittsburgh, PA,
Clone T58.2), TCR-V62-PE (clone B6), TCRVy9- FITC (clone B3), TNF-a-PE-Cy7
(clone
MAB11), Beta Mark TCR Repertoire Kit (Beckman Coulter, Atlanta, GA).
Vector Construction
The polycistronic OSKM vector was previously described (Chang et
al.,"Polycistronic lentiviral vector for "hit and run" reprogramming of adult
skin fibroblasts
to induced pluripotent stem cells," Stem cells 27: 1042-1049 (2009)). The
Lenti-hDL4-
mCherry plasmid was constructed by cloning a PCR-amplified human DL4 cDNA
(Open
Biosystems, LaFayette, CO), an IRES fragment (Open Biosystems) and mCherry
cDNA into
a lentiviral vector (pDL171) which contains the EFla promoter. PCR reactions
were
performed using PrimeStar polymerase (Takara, Mountain View).
To construct CRISPR plasmids, gRNA oligos were designed and introduced into
pX330 and pX335 plasmids following the Zhang lab protocol (Addgene, Cambridge,
MA).
To construct the JAK3 repair plasmid, wild type human genomic DNA was PCR
amplified
using JAK3 primer sets (5' arm:
gtcgacgtcgacgctcagtgaagctgaagtattccttctgcttcacagggcgaccactac (SEQ ID NO: 7)
and
atttaaatcctcccctcgaacccttaccaaactcctatgcatactacag (SEQ ID NO:8); 3' arm:
ttaattaattaattagcattttaggttcaggttgtgagaacactagaagagaacaagtca (SEQ ID NO: 9)
and
gtatacgtatacgcatacctggagaggggacaaggtcttgagatgcgagggt (SEQ ID NO: 10). After
digesting
with enzymes (5' arm: SalI and SwaI; 3' arm: PacI and BstZ17I), the PCR
products were
cloned into a plasmid containing a LoxP-PGK-Neo-LoxP fragment. All of the
oligos used in
this study were synthesized by Integrated DNA Technologies (IDT, Coralville,
IA). To
construct the BCL2 lentiviral plasmid, a primer set (forward:
agccaccttaattaagccaccatggcgcacgctgggagaacggggtacgata (SEQ ID NO: 11) and
reverse:
taacagagagaagttcgtggctccggatcccttgtggcccagataggcacccagggtgat (SEQ ID NO: 12))
was used
to amplify the human BCL2 cDNA (Open Biosystems) fragment. The product was
linked
with GFP through a 2A sequence by PCR and cloned into the pDL171 vector. gRNA-
F1
caccGTG AGA TAC AGA TAC AGA CA (SEQ ID NO: 13) gRNA-R1 aaacTGT CTG TAT
CTG TAT CTC AC (SEQ ID NO: 14) gRNA-F2 caccgAAT GAT TTG CCT GGA ATG CC
(SEQ ID NO: 14) gRNA-R2 aaacGGC ATT CCA GGC AAA TCA TTc (SEQ ID NO: 15)
24

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
gRNA-F3 caccgCAG CCT AGG CAA AGG CCT GC (SEQ ID NO: 16) gRNA-R3
aaacGCA GGC CTT TGC CTA GGC TGc (SEQ ID NO: 17) gRNA-F4 caccgTGC CAA
CAG AAC TGC CTG AT (SEQ ID NO: 18) gRNA-R4 aaacATC AGG CAG TTC TGT
TGG Cac (SEQ ID NO: 19) gRNA-F5 caccGAC CAG GGT GCA AGT GTG GA (SEQ ID
NO: 20) gRNA-R5 aaacTCC ACA CTT GCA CCC TGG TC (SEQ ID NO: 21) gRNA-F6
caccGCT CCT CAG CCT GGC ATT CA (SEQ ID NO: 22) gRNA-R6 aaacTGA ATG CCA
GGC TGA GGA GC (SEQ ID NO: 23)
Cell culture
IP SCs were cultured on mitomycin C-treated MEFs derived from E14.5 CF-1
embryos in ES cell media consisting of DMEM F-12 supplemented with lx non-
essential
amino acids, 1X penicillin-streptomycin, lx L-glutamine (all from Mediatech,
Corning, NY),
20% KnockOut Serum Replacement (Invitrogen), 2-PME (Sigma) and 5-10 ng/mL bFGF
(Invitrogen). Human primary keratinocytes were cultured in DermaLife K Medium
Complete
Kit (LifeLine Cell Technology, Frederick, MD). 0P9 cells were purchased from
ATCC and
grown in a-MEM medium with 20% FBS and penicillin-streptomycin. 0P9-DL4 cells
were
established by transducing 0P9 cells with a lentivirus containing hDL4 and
mCherry.
Virus Production
For preparation of lentivirus, 101.tg of the lentiviral vector, 2.51.tg of the
envelope
plasmid (pMDG), and 7.5 1.tg of the packaging plasmid (pCMBVdR8.9.1) were co-
transfected
into 5x106 293T cells by Fugene 6 (Roche, Nutley, NJ or Promega, Madison, WI).
Virus-
containing supernatant was collected 2 days after transfection and passed
through a 0.45 1.tm
filter.
Gene targeting
IPSCs were treated with 0.25% trypsin for 5 minutes to generate single cell
suspensions. After washing twice with 1X PBS, 1 to 2 million cells were mixed
with 51.tg of
JAK3 repair plasmid and 5 1.tg of pX330-JAK3 or pX335-JAK3 plasmids for
Nucleofection
(Human Stem Cell Nucleofector Kit, program A-023, Lonza, Alpharetta, GA) and
plating
onto MEFs. Two to four days later, hES medium containing 301.tg/mL of G418 was
added to
the plates to select for drug resistant colonies. The colonies were picked 3
to 4 weeks later
and expanded for genomic DNA extraction. For PCR genotyping, a 5' primer set
(tgctaaagcgcatgctccagact (SEQ ID NO: 24) and gtcttcatctcagggtcggct (SEQ ID NO:
25) and a
3' primer set (cctctctgtgcattatggcag (SEQ ID NO: 26) and gccttctatcgccttcttg
(SEQ ID NO:
27)) were used. To remove the Neo selection marker, hiPSCs were infected with
a Cre-
expressing adenovirus (rAd-Cre-IE).

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
RT-PCR
Total RNA was isolated from in-vitro derived cells with Trizol reagent
(Invitrogen,
Carlsbad, CA). cDNA was synthesized with 0.5 to 2 [ig of total RNA using
Superscript First-
strand Synthesis System (Invitrogen) according to the manufacturer's
instructions. SYBR
Green PCR Master Mix (Life Technologies, Carlsbad, CA) was used for qPCR
according to
the manufacturer's instructions. Primer sets used for qPCR are GAPDH (F:
actcctccacctttgacgct (SEQ ID NO: 28), R: tcccctcttcaagggtctacatg (SEQ ID NO:
29)); PU.1
(F: gtgcaaaatggaagggtttc (SEQ ID NO: 30), R: ggagctccgtgaagttgttc (SEQ ID NO:
31));
GATA3 (F: tgtttcctttcactggccaca (SEQ ID NO: 32), R: aacggcaactggtgaacggta (SEQ
ID NO:
33)); BCL11B (F: ggcgatgccagaatagatgccg (SEQ ID NO: 34), R:
ccaggccacttggctcctctatctccaga (SEQ ID NO: 35)); RAG1 (F:
ccttactgttgagactgcaatatcc (SEQ
ID NO: 36), R: ctgaagtcccagtatatacttcacac (SEQ ID NO: 37)); RAG2 (F:
cccagaagcagtaataatcatcgag (SEQ ID NO: 38), R: atgtgggatgtagtagatcttgc (SEQ ID
NO: 39));
pTa (F: gggtcttacctcagcagttac (SEQ ID NO: 40), R: cctcacacagtgtgacgcag (SEQ ID
NO: 41));
BCL2 (F: gactgagtacctgaaccggc (SEQ ID NO: 42), R: gggccaaactgagcagagtc (SEQ ID
NO:
43)); BAX (F: aagaccagggtggttgggac (SEQ ID NO: 44), R: gtaagaaaaatgcccacgtc
(SEQ ID
NO: 45)); and JAK3 (F: agtcagacgtctggagcttc (SEQ ID NO: 46), R:
gtgagcagtgaaggcatgagtc
(SEQ ID NO: 47)). All values were normalized relative to GAPDH expression.
Whole Genome Sequencing and Analysis
DNA from iPSCs was sheared using a Covaris S2 Focused-ultrasonicator: 130 [EL
samples in microTUBEs were subjected to two 40-second cycles of 10% Duty
Cycle,
Intensity of 4, and 200 Cycles per Burst in Frequency Sweeping Mode. DNA Chip
(DNA
1000 Kit; Agilent Technologies, Santa Clara, CA) analysis using an Agilent
2100
Bioanalyzer indicated an average fragment size of 400 bp. Library preparation
was performed
using an NEBNext Ultra DNA Library Prep Kit for Illumina (NEB #E7370), and the
final
library concentration was determined by qPCR using a KAPA Illumina Library
Quantification Kit (KK4835; KAPA Biosystems, Wilmington, MA) and an Applied
Biosystems ViiA 7 Real-Time PCR System (Life Technologies). Sequencing
clusters were
produced on the flow cell using an Illumina TruSeq PE Cluster Kit v3 ¨ cBot ¨
HS (PE-401-
3001) and an Illumina cBot. WGS was performed using an Illumina TruSeq SBS Kit
v3 ¨ HS
¨ 200 cycles (FC-401-3001) and an Illumina HiSeq 2500 upgrade to generate
2x100 single-
index paired-end reads for bioinformatic analysis. Probable off-target sites
were identified by
aligning the CRISPR/Cas9 guide sequences to the hg19 reference genome using
EMBOSS
fuzznuc software (v6.6Ø0) (Rice et al., "EMBOSS: the European Molecular
Biology Open
26

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
Software Suite," Trends in Genetics : TIG 16: 276-277 (2000)) and allowing for
a maximum
of three mismatches; 1193 sites were predicted for the first guide sequence
(GTGAGATACAGATACAGACA) (SEQ ID NO: 48) and 257 sites for the second guide
sequence (AATGATTTGCCTGGAATGCC) (SEQ ID NO: 49). All of the reads from the
WGS for each sample were mapped to the hg19 reference genome using the BWA
(v0.7.5a)
mem algorithm (Li and Durbin, "Fast and accurate long-read alignment with
Burrows-
Wheeler transform," Bioinformatics 26: 589-595 (2010)) and duplicate reads
were removed
using Picard-tools (v1.100) (http://picard.sourceforge.net). Local realignment
and base
quality re-calibration were performed using GATK (v2.7-2) (McKenna et al.,
"The Genome
Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA
sequencing
data," Genome research 20: 1297-1303 (2010)). Both SNVs and indels were called
using the
GATK HaplotypeCaller. Additionally, SNVs and indels were separately re-
calibrated as
described in GATK Best Practices and quality filters were applied. The
variants from the
reference genome that were common to all four iPSC samples were excluded from
CRISPR/Cas9 off-target analysis. The non-excluded variants were screened using
Bedtools
(v2.17.0) (Quinlan and Hall, "BEDTools: a flexible suite of utilities for
comparing genomic
features," Bioinformatics 26: 841-842 (2010)) to determine if they fell within
the probable
off-target sites. The analysis shows that none of these variants reside in the
off-target sites
and suggests these mutations were randomly accumulated. All of the functional
variants
(excluded and non-excluded) with a low allele frequency (< 1%, dbSNP 138) were
then
annotated using the ANNO VAR software package and screened for known
associations with
diseases in HGMD and ClinVar (v20140902); additionally, all of the hits with a
high CADD
score (CADD >= 20) were also screened for associations with complex diseases
using the
GWAS Catalog and COSMIC (v70). No validated disease-associated variants were
identified in the databases queried. Of particular interest, the JAK3 C1837T
(p.R613X)
mutation was also not validated to associate with a disease, though the SNP
(rs149316157) is
predicted to be significantly deleterius, with a GERP score of 3.85 and a CADD
score
(CADD phred-like score) of 38. Therefore, the JAK3 C1837T variant was
associated for the
first time with a clinical case of SCID.
Accession codes
The WGS data can be accessed at the NCBI SRA database with the accession
number
SRP056149.
27

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
JAK3-deficient human T cells express low levels of BCL2 and die at an early
developmental stage
IPSCs were generated from skin keratinocytes (Chang et al., 2009) of a SCID
patient
homozygous for a C>T nucleotide substitution in exon 14 of the JAK3 gene. This
mutation
replaces a CGA codon (arginine at 613) with a TGA stop codon (p.R613X). As
described
above, the four-month-old patient presented with a T-B+NK- clinical phenotype.
To
determine whether this SCID phenotype can be recapitulated in vitro,
differentiation of
patient-specific iPSCs to T lymphocytes using a two-step 0P9 and 0P9-DL4
system (Chang
et al., 2014) was attempted. JAK3-deficient iPSCs grew at a rate comparable to
control
iPSCs derived from healthy donors, and these iPSCs efficiently differentiated
into CD34+
hematopoietic progenitors (HPs) on 0P9 stromal cell monolayers. However, when
the
JAK3-deficient, iPSC-derived CD34+ HPs were plated on 0P9-DL4 stromal
monolayers, T-
cell differentiation was absent compared to controls (Fig. 1). No CD3+ T cells
or CD3-
CD16+CD56+ NK cells were observed (Fig.1A), and no CD4+CD8+ double positive
(DP),
CD4+ single positive (SP), or CD8+ single positive (SP) T cells were detected
(Fig.1B).
Jak3 knockout (KO) mice have a small thymus due to a block in thymocyte
differentiation at
the CD4-CD8- double negative (DN) stage prior to productive TCR rearrangement.
To
further understand the developmental defects resulting from a JAK3 mutation in
humans, T
lineage commitment and maturation of JAK3-deficient cells compared to normal
JAK3 WT
controls was assayed. IPSC-derived CD34+ cells were plated on 0P9-DL4
monolayers, and
cells were harvested and analyzed for lymphocyte markers at T-cell induction
day (TD) 14,
21, 28 and 35 (Fig. 4A). In normal controls, 1.2 X 107 CD7+ cells (84% of
cells counted in
the lymphoid gate) were generated at TD14 from 1-2 X 106 CD34+ cells. T cell
markers
CD4, CD8, CD3 and TCR af3 were sequentially detected upon T cell maturation.
At TD35,
more than 50% of the population was CD8 SP cells. In JAK3-deficient cells,
only 4.5 X 104
CD7+ cells (38.9% of cells counted in lymphoid gate) were generated at TD14
from 1-2 X
106 CD34+ cells. The number of CD7+ cells decreased during extended culture
and T cell
markers CD3, CD4, CD8 and TCR af3 were not significantly expressed. During the
transition
through early T cell progenitors (ETPs), the CD4-CD8- (DN) to CD4+CD8+ (DP)
stages are
directed by precise activation and repression of specific transcription
factors. In control cells,
the silencing of PU.1 and induction of GATA3 and BCL11B (Fig. 1C) suggest that
these
cells proceed to the onset of T lineage commitment (DN2 to DN3) followed by
TCR
rearrangement. In contrast, in JAK3-deficient cells PU.1 accumulates and GATA3
and
BCL11B levels are reduced (Fig. 1C). These data suggest that human JAK3-
deficient cells
28

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
arrest before or at the DN2 stage, which is similar to the stage at which T
cells die in Jak3
KO mice. Interestingly, human JAK3-deficient cells may express sufficient
RAG1, RAG2
and PTCRA (Fig. 1C) to perform TCR rearrangement, but the cells do not survive
long
enough to proceed to this important developmental stage. These profound
defects in
lymphocyte development of JAK3-deficient cells can be explained by the absence
of IL-7
signaling which plays an important role in lymphoid progenitor survival and
differentiation.
IL-7/JAK3 signaling maintains thymocyte homeostasis by regulating the BCL2
family of
apoptotic regulators. Thymocytes and peripheral T cells from Jak3 KO mice have
a high
apoptotic index in part through selectively elevating BAX, a pro-apoptotic
factor, and by
reducing expression of BCL2, an anti-apoptotic factor. Similarly, in these
studies, an
increase in apoptosis of in vitro-derived human JAK3-deficient cells compared
to controls at
TD10 (9% to 2.2%) and TD17 (7% to 1.9%) (Fig. 2A). Consistent with this
phenotype, BAX
levels were increased and BCL2 levels were reduced in JAK3-deficient cells
compared to
controls (Fig. 2B). Forced expression of Bc12 rescues T, but not B or NK cell
development in
yc¨deficient mice (Kondo et al., Immunity 7: 155-162 (1997)). Transplantation
of Jak3 KO
mice with Bc12-expressing Jak3 KO bone marrow cells also improves peripheral T
cell
numbers (Wen et al., Molecular and cellular biology 21: 678-689 (2001)). To
determine
whether overexpression of BCL2 will rescue T cell developmental defects of
human JAK3-
deficient cells, in vitro-derived, JAK3-deficient CD34+ cells were transduced
with a
lentivirus containing a BCL2- 2A-GFP polycistron driven by EFla promoter.
After
transduction, CD34+ cells were plated on 0P9-DL4 monolayers and assayed for NK
and T
cell markers at TD 28. No CD3-CD16+CD56+ NK cells were found in GFP- (JAK3-;
BCL2
low) or GFP+ cells (JAK3-; BCL2+) (Fig 2C). These findings suggest that BCL2
released the
blockage at the DN stage in JAK3-deficient cells. Interestingly, a second
developmental
arrest was evident at the DP stage; no further differentiation of CD8+CD4+ DP
positive cells
was observed in GFP+ cells (Fig 2C). In summary, the studies described above
demonstrate
that human SCID phenotypes can be recapitulated in vitro with patient-derived
iPSCs. JAK3
deficiency results in proliferative defects in DN thymocytes. Forced
expression of BCL2
enhances survival of DN cells, which further differentiate into DP thymocytes.
Nevertheless,
DP thymocytes fail to mature to SP T cells, and this defect may result from
the absence of
1L7/JAK3 signaling.
29

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
Correction of the JAK3 deficiency in SCID hiPSCs by CRISPR/Cas9 enhanced gene
replacement
To determine whether normal T cell development can be restored in JAK3-
deficient
SCID patient cells, the JAK3 mutation was corrected in iPSCs by CRISPR/Cas9
enhanced
gene replacement. Six guide RNAs within introns upstream and downstream of
exon 14
were designed to target wtCas9 or nCas9 near the C1837T mutation, and a
correction
template was used for gene replacement (Fig. 3A). IPSCs were nucleofected with
two
plasmids expressing the DlOA Cas9 nickase and paired guide RNAs or a single
plasmid
expressing wild-type Cas9 and a single guide RNA. Cells were grown in medium
containing
G418 for 2 weeks post nucleofection. Individual colonies were picked,
expanded, and
genotyped by PCR (Fig. 3B Top). The efficiency of CRISPR/Cas9¨mediated JAK3
gene
correction is shown in Fig. 3C. Three clones from WT Cas9 + gRNA #1, 3 clones
from WT
Cas9 + gRNA #2 and 6 clones from Cas9 nickase + paired gRNAs #1 and #2 were
further
verified by Sanger sequencing. In 12 sequenced clones, 2 homozygous corrected
clones (1
clone from Cas9 nickase + paired gRNA #1 and #2, and 1 clone from WT Cas9 +
gRNA #1)
and 10 heterozygous corrected clones were identified (Fig. 3D). Restoration of
JAK3 gene
expression was demonstrated by RT-PCR (JAK3 mRNA) (Fig. 3B; lower left panel)
and
western blot (JAK3 protein) (Fig. 3B; lower right).
Specificity of CRISPR/Cas9 directed JAK3 correction
The potential for off-target, CRISPR/Cas9 directed genome modifications raises
some
concerns about the use of this approach for therapy in humans. In cancer cell
lines, relatively
high levels of off-target mutagenesis by Cas9-gRNAs have been described. To
determine the
specificity of CRISPR/Cas9 directed JAK3 correction in human SCID iPSCs, Whole
genome
sequencing was performed before and after gene replacement. The genomes of two
heterozygous and one homozygous corrected clones were sequenced. The two
heterozygous
clones were corrected with gRNA #2 + wild type Cas9, and the homozygous clone
was
corrected with gRNA #1 + gRNA #2 + nickase Cas9 (D10A). The 20-base CRISPR
guide
sequences were mapped to the human reference genome, allowing up to 3
mismatches in
order to identify possible off-target sites. These sites were then analyzed
for variations in the
iPSC samples following CRISPR/Cas9 directed gene replacement. WGS analysis of
one
homozygous and two heterozygous corrected iPSC lines demonstrated that no
mutations
(SNVs nor indels) were introduced into the predicted off-target sites,
suggesting a strong
specificity for the CRISPR/Cas9 directed gene replacement.

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
Restoration of T cell development after CRISPR/Cas9 directed JAK3 correction
To determine whether T cell development is restored after JAK3 gene
correction, T
cell lineage commitment and maturation were assayed. T cell differentiation
sequentially
passes through intermediates observed in vivo: CD34+CD7+ T/NK committed stage;
CD7+CD4+CD8¨ immature, SP stage; CD4+CD8+ DP stage; and finally, CD3+CD8+
TCRc43 mature stage. Mature T cells are polyclonal, proliferate, and secrete
cytokines in
response to mitogens. Therefore, JAK3 corrected hiPSCs were differentiated
into
hematopoietic progenitors on 0P9 monolayers, and CD34+ cells were positively
selected on
anti-CD34 magnetic beads. These cells were plated on 0P9-DL4 monolayers, and
nonadherent cells were analyzed for lymphocyte markers at TD14, 21, 28 and 35
(Fig. 4).
Similar to control cells, 1-2 X 106 CD34+ JAK3 corrected cells differentiated
into 4.7 X
106 CD7+ cells (91% of cells counted in lymphoid gate) at TD14. After further
differentiation to TD21, TD28 and TD35, T cell maturation markers CD3, CD4,
CD8 and
TCR af3 were abundantly observed (Fig. 4A). To determine whether TCR
rearrangement is
reestablished in JAK3-corrected T cells, TCR VP typing was performed by flow
cytometry
and summarized in Figure 4B. JAK3-corrected T cells expressed all the VP
segments that we
tested (19 of 25); therefore, a broad TCR repertoire was restored. Finally,
the integrity of the
TCR signaling pathway, a surrogate of T cell function, in JAK3-corrected T
cells, was
examined by measuring cell surface activation markers following anti-CD3/CD28
stimulation. On Day 3 post-stimulation, the percentage of CD3+CD25+CD69+ T
cells
increased from 0.68% to 59.7% in JAK3-corrected T cells similar to the
increase observed in
control cells (0.01% to 37.6%) (Fig. 4C). These data and results described
above demonstrate
that correction of the JAK3 C1837T (p.R613X) mutation by CRISPR/Cas9 enhanced
gene
replacement in an in vitro iPSC model system restores normal T cell
development with the
capacity to produce functional, mature T cell populations with a broad TCR
repertoire.
In humans, the phenotype of lymphocytes in the peripheral blood of SCID
patients
has been well described, but studies on critical steps of lymphoid commitment
and
thymocyte development have been difficult to perform. Access to bone marrow
and
thymocyte samples from untreated patients with SCID is challenging since these
conditions
are rare and infants typically present with life-threatening infections
requiring urgent HSC
transplantation to survive. The strategy described herein for studying human
SCID bypasses
these restrictions; large numbers of hematopoietic progenitors can be produced
from patient
specific iPSCs in vitro, and the mechanisms responsible for immunodeficiency
can be
precisely determined. Demonstrated herein is that T cell development in human
JAK3-
31

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
deficient SCID is completely blocked before or at the CD4-CD8- (DN2) stage.
Interestingly,
forced expression of BCL2 enhances survival of DN cells, which further
differentiate into
DP thymocytes. However, DP thymocytes fail to mature to SP T cells, and this
defect may
result from the absence of IL7/JAK3 signaling. It is also demonstrated that
correction of the
human JAK3 mutation by CRISPR/Cas9 enhanced gene replacement restores the
differentiation potential of early T cell progenitors. Corrected progenitors
are capable of
producing NK cells and mature T cell populations expressing a broad TCR
repertoire.
Whole-genome sequencing analysis of one homozygous and two heterozygous
corrected
iPSC lines demonstrates that no mutations (SNVs nor indels) are introduced
into the
predicted off-target sites, suggesting a strong specificity for the
CRISPR/Cas9 directed gene
replacement.
In the methods described herein, CD34+ HSCs can be generated from hiPSCs by co-
culturing with human bone marrow stromal stem (hMSC) cells (See Figure 5). The
HSCs
produced by this method from patient-specific iPSC after gene
correction/modification could
be transplanted back into the patient to treat diseases such as sickle cell
disease (SCD), SCID
or cancer. In the methods described herein, T cells can be generated by
culturing hiPSC
derived CD34+ cells by co-culturing the hiPSC derived CD34+ cells with hMSC-
DL4 (See
Figure 6). HSCs produced by this method from patient-specific iPSC after
correction/modification could be transplanted back into the patient to treat
diseases. The T
cells can comprise y6 T cells. As shown in Figure 7, y6 T cells expressing
recombinant T cell
receptor (TCR) can be efficiently produced from genetically modified iPSC.
Production of
y6 T cells expressing TCR specific for tumor antigens provide a cellular
therapy for cancer.
Example 2
Correction of a mutation associated with Sickle Cell Anemia
by CRISPR/Cas9 enhanced gene replacement
Vector Construction
The human codon optimized S. pyogenes Cas9 with both N-terminal and C-terminal
nuclear localization sequences (nls-Cas9-n1s) were PCR cloned from px330
vector (Addgene
ID: 42230) into a modified pET-28b (EMD Biosciences) vector with a His6-SUMO
tag at the
N-terminus. A gene block cassette containing a short linker peptide followed
by a
supercharged GFP with a net charge of +36 and a 23 amino acid influenza virus
hemagglutinin HA-2 variant peptide INF7 (GLFEAIEGFIENGWEGMIDGWYG)(SEQ ID
NO: 50) was codon optimized for E. coil and synthesized (IDT DNA) and cloned
to fuse with
32

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
the C-terminus of the nls-Cas9-nls. An HIV-TAT peptide (YGRKKRRQRRRPPQ) )(SEQ
ID
NO: 51) coding sequence was also synthesized (IDT DNA) and cloned to fuse with
the N-
terminus of the nls-Cas9-nls.
Protein Overexpression and Purification
The pET-SUMO-scCas9 plasmid was transformed into E. coil strain RosettaTM
2(DE3) cells (EMD Millipore, Billerica, MA) in LB medium. The cells were grown
at 37 C
until the optical density reached 0.6 at 600nm. Induction of protein
overexpression was
achieved by adding 0.5 mM isopropyl-l-thio-B-D-galactopyranoside (IPTG) and
culturing
overnight at 18 C in a shaker. The harvested cells were re-suspended in Ni-
binding buffer
(20mM Tris-HC1 pH 8.0, 1.5 M NaC1, 25 mM imidazole and 0.2 mM TCEP) and lysed
by
Emulsiflex C3 high pressure homogenizer (Avestin). Polyethyleneimine (PEI)
with final
concentration of 0.4% was added into the cleared lysate to precipitate the
nucleic acids. The
proteins in the supernatant after centrifugation was then precipitated by
ammonium sulfate to
remove the PEI and re-dissolved in the Ni-binding buffer. The proteins were
first purified by
a HisTrap nickel affinity column (GE Healthcare) followed by overnight
digestion with
SUMO protease Ulp I at 4 C. The cleaved His-SUMO tag was then removed via a
second
HisTrap column. The flow though containing the scCas9 protein was diluted to
reach the
final NaC1 concentration of 0.5 M and purified on a HiTrap Heparin column (GE
Healthcare)
by gradient elution with buffer containing 20mM Tris-HC1 pH 8.0, 2.0 M NaC1,
and 0.2 mM
TCEP. The eluted scCas9 protein was further purified by a size exclusion
column Superdex
200 16/600 (GE Healthcare) in gel filtration buffer (20mM Tris-HC1 pH 8.0, 0.5
M NaC1, and
0.2 mM TCEP), sterilized by passing through a 0.22 p.m filter and concentrated
by an
Amicon Centrifugal Unit (EMD Millipore) with 100 kDa cutoff. The concentrated
protein
was quantified by UV spectrophotometer and flash frozen in liquid nitrogen.
Guide RNA Preparation
Template DNA for sgRNA transcription was generated by PCR with primer set
adding a T7 promoter and a polyA sequences. sgRNA was in vitro transcribed by
T7 RNA
polymerase using T7 Ribomax Express System (Promega, Madison, WI) according to
the
manufacturer's manual. The transcribed RNA was purified by phenol: chloroform
extraction,
ethanol precipitation and followed by column purification with MEGAc1earTM
Transcription
Clean-Up Kit (Ambion, Austin, TX). The purified gRNA was quantified by UV
spectrophotometer and stored in -80 C freezer.
Single-stranded DNA Donors
Single-stranded DNA (ssODN) donors were synthesized by IDT DNA.
33

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
Single-stranded Donor DNAs for HBB sickle correction
HBB-T2-ssODN ATCCACGTTCACCTTGCCCCACAGGGCAGTAACGGCAGACTTCT
CCtCAGGAGTCAGGTGCACCATGGTGTCTGTTTGAGGTTGCTAGT
GA (SEQ ID NO: 52)
HBB-T2-ssODN- CTTCATCCACGTTCACCTTGCCCCACAGGGCAGTAACGGCAGAtT
wobble TtTCCtCAGGAGTCAGGTGCACCATGGTGTCTGTTTGAGGTTGCT
AGTGA (SEQ ID NO: 53)
Cell Culture
Human sickle patient iPSC were derived from skin fibroblasts and were
maintained
on Matrigel (BD) in mTeSRTml medium (Stem Cell Technologies, Vancouver, CA)
with
penicillin/streptomycin.
scCas9-sgRNA-ssODN complex preparation and nucleofection
1/10 volume of 10x PBS was added into sgRNA to reach lx final concentration.
The
sgRNA was annealed on PCR thermo cycler with slow decreasing of temperature
from 95 C
to 4 C. After annealing, scCas9 protein was added into the sgRNA with a 1:1.5
protein-to-
RNA molar ratio and mixed quickly by tapping the tube until all the transient
precipitation
was gone. The mixture was incubated in room temperature for 10 minutes in
dark. Then, 1
molar ratio amount of ssODN was added into the mixture and incubated for
additional 10
minutes in dark to form the scCas9-sgRNA-ssODN complex.
One day before nucleofection, cells were detached by Accutase (Stem Cell
Technologies) and 1x106 cells /well cells were seeded on a 6-well plate with
10 i.tM Rock
inhibitor (Y-27632) (EMD Millipore). For each experiment, 5x105 hsIPSCs were
resuspended as single cells in 100 1 supplemented Human Stem Cell Nucleofector
Solution
l(Lonza) and scCas9-sgRNA-ssODN complex was then mixed with the cell solution.
The
cells were nucleofected with program A-023 using a Nucleofector II device
(Lonza, Basel,
Switzerland). The efficiency of HBB genome correction was analyzed by ddPCR
two days
post nucleofection.
Detection of sickle correction by ddPCR
The cells nucleofected with the scCas9-sgRNA-ssODN complex were lysed by
prepGEM Tissue DNA extraction reagent (ZyGEM, Hamilton, NZ) following
manufacturer's
manual and 1:3 diluted with water. In a 22 11.1 ddPCR reaction, 11 11.1 2 x
ddPCR mix (Bio-
rad) was mixed with lul each of 5 M allele-specific FAM or VIC Taqman probes
set forth
34

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
below, 0.2 ul each of a 100 uM forward and reverse primer, and 8.6 ul diluted
genomic
DNA. Droplets were generated by QX200 Droplet Generator (Bio-rad, Hercules,
CA)
according to the manufacturer's manual. The reaction mix was then transferred
into a 96-well
PCR plate and the PCR was performed on a standard thermal cycler (Bio-rad).
The program
for PCR was : Step 1: 95 C 10 min; Step 2: 95 C 30s; Step 3: 55 C 1 min;
repeat steps 2-3
for 39 times; Step 4: 98 C 10 min; Step 5: 8 C hold. After PCR was done, the
plate was then
analyzed by QX200 Droplet Reader (Bio-rad).
T7-sgRNA transcription template primers
T7-T2-F TAATACGACTCACTATAGGGTAACGGCAGACTTCTCCAC (SEQ ID NO: 54)
T7- AAAAAGCACCGACTCGGTGCC (SEQ ID NO: 55)
polyA-R
Taqman Probes:
HBB-wb-FAM-TM FAM-TCCTGaGGAaAAaT-MGB (SEQ ID NO: 56)
HBB-wt-FAM-TM FAM-TGACTCCTGAGGAGAA-MGB (SEQ ID NO: 57)
HBB-sk-VIC-TM VIC-ACTCCTGTGGAGAAG-MGB (SEQ ID NO: 58)
ddPCR Primers:
R196 HBB-TaqM-f2 CAGAGCCATCTATTGCTTACATTTG (SEQ ID NO: 59)
R197 HBB-TaqM-r1 GGCCTCACCACCAACTTCAT (SEQ ID NO: 60)
As set forth above, a complex that includes a guide RNA (gRNA), modified
recombinant Cas9 protein (mrCas9) and a single-stranded
oligodeoxyribonucleotide
(ssODN) can be introduced into human stem cells or derivatives thereof to
correct a single
base mutation that causes disease. Table 1 and Figure 8 illustrate results
from the
introduction of a sickle cell correction complex (gRNA-mrCas9-ssODN) into
induced
Pluripotent Stem Cells (iPSC) derived from skin cells of a sickle cell
patient. IPSCs were
derived as described in Example 1. The correction complex was introduced into
sickle iPSC
by nucleoporation and 2 days later genomic DNA was analyzed by digital PCR,
using the
primers set forth above, and sequenced. Over 65% of the cells contained at
least one

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
corrected gene. One corrected gene is sufficient to cure the disease. The
results were
confirmed as follows. Two days after introduction of the correction complex,
the cells were
plated in culture dishes, and 43 individual iPSC colonies were isolated.
Genomic DNA was
isolated from these colonies and the beta-globin gene was sequenced. Sixty-
five percent of
the colonies contained at least one corrected beta-globin gene (S corrected to
A).
Table 1
................................................. ilititklorCooliwssODN
Pooled thiPCR fintfit (2-eley) =
Total aolonies oftor 2 wedts
............................
:::.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.............:.:.:
.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.:.
IteitiXed totortiet
_________________________ Total sink tolonies ........ 4:3
..............
A/A14
AI 4
ladetliodel ... 6
=== ::::::::
etOMS With ttt kW*. oilole otowsdod .. 651%
......
= ====
Cionito with Witht1 i
tionct with room* mothikation ........
86
mmAr4t&l=raitciogumiikuir:::M
µ,µ,%µw ............................................................
Similar studies were performed with patient primary bone marrow CD34+ cells.
The
protocol was as follows. Bone marrow was obtained from a sickle patient by an
MB
approved protocol. CD34+ cells were purified on a Miltenyi anti-CD34+ beads
(Miltenyi,
Bergisch Gladbach, Germany). The cells were nucleoporated with the complex
prepared as
described above. After nucleoporation, the cells plated in methycult and BFU-
E, CFU-E and
CFU-GEMM colonies were picked after two weeks and analyzed for corrected
alleles. Table
2 and Figure 9 illustrate results from the introduction of a sickle cell
correction complex
(gRNA-mrCas9-ssODN) into patient primary bone marrow CD34+ cells. After twelve
days
of in vitro differentiation, DNA was analyzed by digital PCR (ddPCR) and
sequenced.
36

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
Approximately equal amounts of betaA and betaS mRNA were observed (See Figure
9).
Immediately after nucleoporation, some of the cells were culture in erythroid
differentiation
medium for up to eightenn days and enucleated red blood cells were analyzed
for HbA. An
isoelectric focusing (IEF) gel of in vitro differentiated red blood cells from
the corrected
sickle patient CD34+ cells showed an HbA (normal hemoglobin) to HbS
(hemoglobin with
sickle cell mutation) ratio of about 1:3, which is sufficient to inhibit
sickling and treat the
disease (See Figure 10).
Table 2
Complex for nucleofection Cas9wt-36GFP-T2-ssODN
Nucleofection Program P4 DN-100
..........................................................
BFU-E/M-E/GEMM colonies picked on D10 and 015 .21/23/7
...........
:=...: ..k A/A :2 4
,...:.===================..... ================================
:=:::=:=:::
Indeliindel
c191.1.. W.411 OOP corrected4
...........
; = = = = = = = = = = = = = = = = =
-.. im 'im: ; = = = = = = = = = = =:. 1 ; =
= = = = = = = = = i:nrm
aones with indel5
........................................................................
...:.:.:.:.:.:.:.:.:.:.:
Clones with genome modification
EgggggggggggT6t4liiii4kiliefiififfelOggEMMEMMg ggggggggggmliammommgm?.,
iggggIggggl. Megggl
mmmmmmmmmmmmmmmmmmmmmmmu::mmmmmomnmmmm
37

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
Example 3
Correction of a mutation associated with Sickle Cell Anemia
by CRISPR/Cas9 enhanced gene replacement
iPSCs have the potential to generate all cell types including HSPCs (human
stem/progenitor cells); therefore, iPSC based gene therapy could provide a
curative therapy
for sickle cell disease. Correction of sickle iPSCs can provide an unlimited
number of cells
from which to generate corrected HSPCs, and these corrected HSPCs can be used
for
autologous transplantation. Importantly, corrected iPSCs and the HSPCs derived
from them
can be fully characterized and evaluated for safety before transplantation.
Described below is
CRISPR/Cas9 enhanced gene correction of iPSCs derived from fibroblasts of a
sickle patient.
Cell Culture
Human sickle iPSCs
Human sickle iPSCs were derived from fibroblasts of a skin biopsy obtained
from a
consented sickle patient at the UAB Kirklin Clinic. The cells were maintained
on Matrigel
(BD) in mTeSRTml medium (Stem Cell Technologies) with penicillin/streptomycin.
Human
sickle iPSCs were passaged every 3-4 days by incubating colonies with Accutase
(Stem Cell
Technologies), and single cells were seeded on Matrigel coated plates with 10
i.tM Rock
inhibitor (Y-27632) (EMD Millipore). After one day, the media was changed with
no rock
inhibitor.
Human sickle bone marrow CD34+ cells
Bone marrow from a consented sickle patient was aspirated in the adult sickle
clinic at
UAB. The CD34+ cells were purified on anti-Cd34+ beads, aliquoted and stored
in liquid
nitrogen.
Cas9 Expression Plasmids for E. coli overexpression
Cas9WT
The S. pyogenes Cas9WT coding sequence with both N-terminal and C-terminal
fused
nuclear localization sequences (nls-Cas9WT-n1s) were PCR cloned from the px330
vector
(Addgene ID: 42230) into a modified pET-28b (EMD Biosciences) vector with a
His6-SUMO
tag at the N-terminus, resulting in a pSUMO-Cas9WT plasmid.
38

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
TAT-Cas9WT-EGFP
Synthesized genes block (IDT DNA) containing a short linker peptide and the
coding
region of EGFP were ligated to the C-terminus of the nls-Cas9WT-nls and
cloned. Coding
sequence for a HIV-TAT peptide (YGRKKRRQRRRPPQ)(SEQ ID NO: 51) was also
synthesized, ligated to the N-terminus of the nls-Cas9WT-nls and cloned,
resulting in the
pSUMO-TAT-Cas9WT-EGFP plasmid.
Cas9WT-36GFP
A synthesized gene block (IDT DNA) containing the E. coil codon optimized
coding
sequence of supercharged GFP with a net positive charge of +36 (Lawrence et
al.
"Supercharging Proteins Can Impart Unusual Resilience," I Am. Chem. Soc.
129(33): 10110
(2007))) and short linker peptide was ligated with the C-terminus of the nls-
Cas9WT-nls and
cloned, resulting in a pSUMO-Cas9WT-36GFP plasmid.
TAT-Cas9WT-36GFP
The coding sequence of a HIV-TAT peptide (YGRKKRRQRRRPPQ)(SEQ ID NO:
51) was synthesized, ligated with the C-terminus of Cas9WT-36GFP and cloned,
resulting in
the pSUMO-TAT-Cas9WT-36GFP vector.
TAT-Cas9WT-36GFP-INF7
A synthesized gene block (IDT DNA) containing a short linker peptide followed
by a
supercharged GFP with a net charge of +36 (Lawrence, 2007) and a 23 amino acid
influenza
virus hemagglutinin HA-2 variant peptide INF7 (GLFEAIEGFIENGWEGMIDGWYG)(SEQ
ID NO: 50) (Plank, 1994) was codon optimized for E. coil, ligated with the C-
terminus of the
nls-Cas9WT-nls and cloned. An HIV-TAT peptide (YGRKKRRQRRRPPQ)(SEQ ID NO:
51) coding sequence was also synthesized, ligated with the N-terminus of nls-
Cas9-nls and
cloned, resulting in the pSUMO-TAT-Cas9WT-36GFP-INF7 plasmid.
Cas9WT-3xTAT
The coding sequence of 3 tandem repeats of the coding region for HIV-TAT
peptide
separated with short linkers
(YGRKKRRQRRRPPQAGGGSGGSYGRKKRRQRRRPPQAGGGSGGSYGRKKRRQRR
RPPQAG) (SEQ ID NO: 61) was codon optimized for E.coli, synthesized, ligated
with the C-
terminus of nls-Cas9WT-nls and cloned, resulting in the pSUMO-Cas9WT-3xTAT
plasmid.
TAT-Cas9WT-3xTAT
The coding sequence of a HIV-TAT peptide was (YGRKKRRQRRRPPQ)(SEQ ID
NO: 51) synthesized, ligated with the N-terminus of nls-Cas9WT-3xTAT and
cloned,
resulting in a pSUMO-TAT-Cas9WT-3xTAT plasmid.
39

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
Protein Overexpression and Purification
The Cas9WT or Engineered positively charged Cas9 (EpcCas9) expression plasmid
was transformed into the E. coil strain RosettaTM 2(DE3) cells (EMD Millipore)
in LB
medium. The cells were grown at 37 C until the optical density reached 0.6 at
600nm.
Induction of protein overexpression was achieved by adding 0.5 mM isopropyl-l-
thio-B-D-
galactopyranoside (IPTG) and culturing overnight at 18 C in a shaker
incubator. The
harvested cells were re-suspended in Ni-binding buffer (20mM Tris-HC1 pH 8.0,
1.5 M NaC1,
25 mM imidazole and 0.2 mM TCEP) and lysed with a Emulsiflex C3 high pressure
homogenizer (Avestin). Polyethyleneimine (PEI) was added to the cleared lysate
supernatant
to a final concentration of 0.4% to precipitate nucleic acids. The supernatant
after
centrifugation was then precipitated by ammonium sulfate to remove the PEI and
the protein
pellet was re-dissolved in the Ni-binding buffer. The protein solution was
first purified by a
HisTrap nickel affinity column (GE Healthcare, Atlanta, GA) followed by
overnight
digestion with SUMO protease Ulp I at 4 C. The cleaved His-SUMO tag was then
removed
by passing through a second HisTrap column. The flow through containing the
Cas9 protein
was diluted to reach a final NaC1 concentration of 0.5 M and purified on a
HiTrap Heparin
column (GE Healthcare) by gradient elution with buffer containing 20mM Tris-
HC1 pH 8.0,
2.0 M NaC1, and 0.2 mM TCEP. The eluted Cas9 protein was further purified by a
size
exclusion column Superdex 200 16/600 (GE Healthcare) in gel filtration buffer
(20mM Tris-
HC1 pH 8.0, 0.5 M NaC1, and 0.2 mM TCEP), sterilized by passing through a 0.22
p.m filter
and concentrated by an Amicon Centrifugal Unit (EMD Millipore) with a 100 kDa
cutoff.
The concentrated protein was quantified by UV spectrophotometer, flash frozen
in liquid
nitrogen and stored at -80 C.
Single Guide RNA Preparation
The DNA template for sgRNA in vitro transcription was generated by PCR with
primers adding a T7 promoter at 5' end and a polyA sequence at the 3' end. The
sgRNAs was
in vitro transcribed by T7 RNA polymerase using a T7 Ribomax Express Kit
(Promega)
according to the manufacturer's manual. The transcribed RNA was then isolated
by phenol:
chloroform extraction, ethanol precipitation and column purification with the
MEGAc1earTM
Transcription Clean-Up Kit (Ambion). The sgRNA was eluted in nuclease free
water, and the
concentration was measured by UV spectrophotometer. The stock sgRNA was then
aliquoted
and stored in a -80 C freezer.

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
Cas9 RNP/ssODN Assembly
Before complexing with Cas9 protein, 10x PBS was added into the stock sgRNA
solution to reach lx PBS final salt concentration. The sgRNA was annealed on a
thermo-
cycler by slowly decreasing the temperature from 95 C to 4 C. To form Cas9
RNP, stock
Cas9 protein was added to the annealed sgRNA at a 1:1.5 protein:RNA molar
ratio and mixed
thoroughly by quickly tapping the tube until all the transient precipitation
was gone. The
mixture was incubated at room temperature for 10 minutes in the dark.
Subsequently, ssODN
was added at a 1:1 molar ratio with Cas9 RNP for nucleoporation.
Nucleoporation of human sickle iPSCs with Cas9 RNP/ssODN
One day before nucleoporation, human sickle iPSCs were detached by accutase
(Stem
Cell Technologies) and incubated to obtain a single cell suspension in mTesR1
media
supplemented with 10 tM Rock inhibitor (Stem Cell Technologies). This single
cell
suspension was seeded into 6-well plate at density of 5x105 cells/well. On the
day of
nucleoporation, 5x105 human sickle iPSC cells were prepared with Accutase as
described
above and resuspended in 10011.1 of Human Stem Cell Nucleofector Solution 1
(Lonza) and
7.5 tM of Cas9RNP/ssODN was mixed with the cell suspension in the
nucleoporation
cuvette. The cells were nucleoporated with program A-023 using a Nucleofector
II (Lonza)
and transferred into pre-warmed media immediately. The correction efficiencies
for the cell
population were assayed 2 days after nucleoporation.
Detection of sickle correction by ddPCR
Two to five days after nucleoporation, Cas9 RNP/ssODN nucleoporated cells were
lysed by prepGEM Tissue DNA extraction reagent (ZyGEM) following the
manufacturer's
manual and the cell lysate was diluted 1:3 with water. In a 22 11.1 ddPCR
reaction, 11 11.1 2 x
ddPCR mix (Bio-Rad) was mixed with lul each of 5 M allele-specific FAM or VIC
Taqman
probes, 0.2 11.1 each of a 100 forward and reverse primer, and 8.6 11.1
diluted cell lysate.
Droplets were generated by a QX200 Droplet Generator (Bio-Rad) according to
the
manufacturer's instructions. The reaction mix was then transferred into a 96-
well PCR plate,
and PCR was performed on a standard thermal cycler (Bio-Rad). The program for
PCR was:
Step 1: 95 C 10 min; Step 2: 95 C 30s; Step 3: 55 C lmin; repeat steps 2-3 for
39 times;
Step 4: 98 C 10min; Step 5: 8 C hold. After PCR was completed, the plate was
analyzed on
a QX200 Droplet Reader (Bio-Rad).
Generation of single iPSC clone after Cas9 RNP/ssODN nucleoporation
To generate single iPSC clones, Cas9 RNP/ssODN nucleoporated sickle iPSCs were
seeded in BD matrix gel coated 96-well plates after serial dilution to a
density of 20, 10 and 5
41

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
cells/well. Fresh mTesR1 media with 10 i.tM rock inhibitor was changed every 2
days during
the first 6 days of culture. mTesR1 media without rock inhibitor was changed
every day after
day 6. Ten to twelve days after seeding, single iPSC colonies were picked, and
the cell
lysates were analyzed by Sanger sequencing for genome modification.
Activation and Nucleoporation of human patient bone marrow sickle CD34+ cells
To activate the cell cycle, frozen human sickle bone marrow CD34+ cells were
thawed and resuspended into pre-warmed Stemspan media supplemented with CC110
cytokine cocktail (STEMCELL Technology). The cells were cultured in a 37 C
incubator
with 5% CO2 and fresh media was partially changed every day for 2 days before
nucleoporation. On the day of nucleoporation, 5x105 liveCD34+ cells were
rinsed with
1xPBS and harvested by centrifugation at 150g for 15 mins. The cell pellet was
resuspended
in 10011.1 P4 primary cell nucleofection solution (Lonza) and 15 i.tM of Cas9
RNP/ssODN
complex was mixed with the cell suspension in the nucleoporation cuvette. The
cells were
nucleoporated with program DN-100 using a 4D-Nucleofector (Lonza) and
transferred into
pre-warmed media immediately. The efficiency of gene correction was analyzed 6
days after
nucleoporation.
Erythroid Colony forming Unit (CFU) assay for Cas9 RNP nucleoporated CD34+
cells
After nucleoporation with Cas9 RNP/ssODN complex, CD34+ cells were seeded into
Methocult media (Stem Cell Technologies) at a density of 500-1000 cells/mL in
35mm tissue
culture plates. Cells were grown in a 37 C incubator with 5% CO2 for 12-15
days until the
colonies were large enough to pick individually for analysis.
In vitro erythroid differentiation of CD34+ HSPCs into RBCs
One day after the nucleoporation of CD34+ cells with Cas9 RNP/ssODN complex,
the media was changed to Erythroid expansion media (Stemspan SFEM (STEMCELL
Technologies) supplemented with lu/mL erythropoietin (EPO), 2 nM dexamethasone
(DEX),
1nM 13-Estradiol, 2Ong /mL human SCF, and 5ng /mL human IL-3.) The media was
changed
every 2 days. After the first 7 days of expansion and differentiation, the
media were
supplemented with a higher concentration of EPO (2 u/mL) until differentiated
RBCs are
harvested at day 15-18.
Mass spectrometry analysis of corrected hemoglobin beta protein in RBCs
Hemolysates of RBCs differentiated from human sickle bone marrow CD34+ HSPCs
were separated by PAGE. The globin band was cut out of the gel and
trypsinized. Peptides
were separated and analyzed by LC-MS/MS.
42

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
Sequences
In vitro transcribed sgRNA sequences:
Ti sgRNA:
GGGUCUGCCGUUACUGCCCUGGUUUUAGAGCUAGAAAUAGCAAGUUAAAAUA
GGUGCUUUUUUU
(SEQ ID NO: 62)
T2 sgRNA:
GGGUAACGGCAGACUUCUCCACGUUUUAGAGCUAGAAAUAGCAAGUUAAAAU
GGUGCUUUUUU
U (SEQ ID NO: 63)
91-nt correction ssODN:
ATCCACGTTCACCTTGCCCCACAGGGCAGTAACGGCAGACTTCTCCtCAGGAGTC
AGGTGCACCATGGTGTCTGTTTGAGGTTGCTAGTGA (SEQ ID NO: 52)
95-nt T2 wobble ssODN:
CTTCATCCACGTTCACCTTGCCCCACAGGGCAGTAACGGCAGAtTTtTCCtCAGGAG
TCAGGTGCACCATGGTGTCTGTTTGAGGTTGCTAGTGA (SEQ ID NO: 53)
90-nt Ti wobble ssODN:
ACCTCAAACAGACACCATGGTGCACCTGACTCCTGAGGAGAAGTCTGCCGTCAC
AGCTCTGTGGGGCAAGGTGAACGTGGATGAAGTTGG (SEQ ID NO: 64)
PCR primers for T2 sgRNA in vitro transcription template
T7-T2F: TAATACGACTCACTATAGGGTAACGGCAGACTTCTCCAC (SEQ ID NO: 54)
T7-R: AAAAAGCACCGACTCGGTGCC (SEQ ID NO: 55)
PCR primers for Ti sgRNA in vitro transcription template
T7-T1F: TAATACGACTCACTATAGGGTCTGCCGTTACTGCCCTG (SEQ ID NO: 65)
T7-R: AAAAAGCACCGACTCGGTGCC (SEQ ID NO: 55)
PCR primer for on-target Sanger sequencing
R157: TCCACATGCCCAGTTTCTAT (SEQ ID NO: 66)
R158: AGTAGCAATTTGTACTGATGGTATG (SEQ ID NO: 67)
Engineered positively charged Cas9 RNPs/ssODN (EpcCas9 RNPs/ssODN) efficiently
correct the sickle mutation in human patient iPSC (induced Pluripotent Stem
Cells)
To correct the sickle HBB gene, human sickle patient derived iPSCs were
nucleoporated with Cas9WT/T2 RNP, Cas9WT-EGFP/T2 or 8 different EpcCas9/T2
RNPs
(Engineered positively-charged Cas9/T2 RNPs) together with a 91-nt ssODN
correction
43

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
template (SEQ ID NO: 51). Cas9/T2 RNPs induce a double strand break near (2 bp
downstream) the sickle mutation. The proximity of the cut site to the mutation
enhances
HDR of the sickle mutation (T->A) using the 91-nt ssODN correction template.
On-target
Sanger sequencing data for the population of iPSCs demonstrate correction of
the sickle
mutation at high efficiency in Cas9WT RNP/ssODN nucleoporated cells (Fig 11).
The
addition of an EGFP (Enhanced Green Fluorescent Protein) domain at the C-
terminus of
Cas9WT did not affect the level of correction.
Correction efficiencies vary in cells nucleoporated with the 8 different
EpcCas9s RNPs. The
addition of a positively charged HIV TAT peptide at the N-terminus of the
Cas9WT-EGFP
(TAT-Cas9WT-EGFP) results in a small decrease in correction efficiency
compared to the
Cas9WT and Cas9WT-EGFP and a small decrease in indels. Addition of 3x tandem
repeats
of TAT at the N-term of the Cas9WT-EGFP (3xTAT-Cas9WT-EGFP) almost completely
abolishes correction and indel levels, indicating loss of Cas9 enzymatic
activity from this
modification. This result suggests that a relatively high number of positive
charges linked to
the N-terminus of Cas9 severely inhibits enzymatic activity. Interestingly,
addition of
positive charges at the C-terminus of Cas9 (Cas9WT-3xTAT or Cas9WT-36GFP)
results in a
high level of correction and a relatively low level of indels. These results
suggest that
positive charges linked to the C-terminus of Cas9 significantly inhibit
exonuclease digestion
of cleaved ends and stimulate religation of ends without formation of indels.
Similar levels
of correction and indels were observed from EpcCas9 with a C-terminal addition
of 3x
tandem repeats of TAT peptides or a positively charged +36GFP.
EpcCas9s with both N-terminal and C-terminal positively charged modifications
(TAT-Cas9WT-3xTAT and TAT-Cas9WT-36GFP) produce significantly less indels.
Interestingly, further addition of a negatively charged INF7 peptide to the C-
terminus of
TAT-Cas9WT-36GFP (TAT-Cas9WT-36GFP ¨INF7) significantly enhances the
correction
efficiency compared to TAT-Cas9WT-36GFP. Sanger sequencing results were
verified by
deep sequencing analysis of on-target correction and indels for iPSC
populations after
nucleoporation with Cas9WT and selected EpcCas9 RNPs (Fig. 12).
EpcCas9 RNPs suppress on-target indels in human sickle iPSC
To study further the effects of positively charged modifications on the
efficiency of
HDR based gene corrections and NHEJ based indels, human sickle iPSC were
nucleoporated
with Cas9 RNPs plus or minus a 91-nt ssODN correction template. On-target
Sanger
sequencing analysis demonstrated that addition of ssODN (+ssODN) to both
Cas9WT RNP
and TAT-Cas9WT-EGFP corrects the sickle mutation with a similar high
efficiency (Fig 13).
44

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
However, in the absence of ssODN (-ssODN), indel formation is dramatically
lower with
TAT-Cas9WT-EGFP compared to Cas9WT. Since HDR requires DSBs (Double Strand
Breaks), the enzymatic activity of Cas9 is apparently not lowered by the
addition of 1XTAT.
Therefore, the large discrepancy in indel formation is not due to lower
transduction efficiency
or lower enzymatic activity of TAT-Cas9WT-EGFP.
To confirm these observations, the correction and indel efficiencies in 5
other
EpcCas9 RNPs with (+) or without (-) ssODN (Fig. 14) was evaluated. Sanger
sequencing
analyses confirmed that all EpcCas9 RNPs result in significantly fewer on-
target indels in the
absence of ssODN (-ssODN). Although the correction efficiencies of EpcCas9
RNPs/+ssODN vary with different positively charged modifications (Fig. 14),
indel
formation is suppressed by all positively charged Cas9 modifications.
EpcCas9 RNPs enhance cell survival after nucleoporation in human sickle iPSC
To determine whether positively charged modifications affect cell survival,
sickle
iPSC were nucleoporated with Cas9WT RNP or 7 different EpcCas9s with (+) or
without (-)
a correction ssODN. Immediately after nucleoporation, cells were plated in
culture dishes
and growth was examined after 48 hours. Cell survival was poor with Cas9WT and
increased
dramatically with higher positively charged modifications (Fig. 15). Excellent
cell survival
was achieved with Cas9WT-36GFP and EpcCas9s containing both N-terminal and C-
terminal positively charged modifications (Fig 15). Considering cell survival
and
correction/indel efficiency, the optimum balance of high correction, low indel
formation and
excellent cell survival is achieved with Cas9WT-36GFP and TAT-Cas9-36GFP-INF7
RNPs
in human sickle iPSCs.
ssODN: Cas9 RNP ratios for sickle correction in human iPSC
The ratio of ssODN correction template to Cas9 RNP (ssODN:Cas9 RNP) is
critical
important for HDR and cell survival. Single stranded ODN is toxic to cells;
therefore, high
ssODN:Cas9 RNP ratios may result in poor cell survival after nucleoporation.
However, low
ssODN:Cas9 RNP ratios may result in inefficient HDR. To achieve high
correction
efficiencies with high cell survival, ssODN:Cas9 RNP ratios were optimized.
The efficiency
of sickle mutation correction with increasing doses of ssODN in sickle patient
iPSC was
determined. A Cas9WT-36GFP:T2 sgRNA molar ratio of 1:1.35 was fixed for these
experiments, and the molar ratios of ssODN:Cas9WT-36GFP RNP ranged from 0 to
2.0 (r=
0, 0.2, 0.5, 1.0, 1.15, 1.35, 1.5 and 2.0). For example, the r=0.5 value in
Fig. 6 is 0.5
ssODN:1.0 Cas9WT-36GFP:1.35 T2 sgRNA. Forty-eight hours after nucleoporation
of the
ssODN:Cas9WT-36GFP RNPs, sickle corrections were quantitated by digital
droplet PCR

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
(ddPCR) (Fig. 16A) and Sanger sequencing (Fig. 16B). The percent correction
(betaA/betaS
alleles x 100) was plotted verses r (ssODN:Cas9WT-36GFP RNP). Correction
efficiencies
increased with r=0.2 to r=1.0 and reached a plateau at 1.15 (65.7%).
Increasing r above 1.15
did not significantly increase correction efficiency and dramatically
inhibited cell survival.
Cas9:sgRNA ratios for sickle correction in human iPSC
Theoretically, the optimal Cas9:sgRNA molar ratio is 1:1. Saturation of the
Cas9
protein with sgRNA ensures maximal Cas9 enzymatic activity and reduces the
possibility of
free Cas9 interactions with other small RNAs that may produce unpredictable
off-target
genome modifications. Small RNAs are sensitive to nucleases; therefore, molar
ratios of
Cas9:sgRNA greater than 1:1 may be necessary to saturate Cas9. Cas9-
36GFP:sgRNA molar
ratios of 1:1.15, 1:1.35 and 1:1.5 were tested with ssODN molar ratios of 1.15
or 1.35 to
determine optimal correction efficiency of the sickle mutation in patient
iPSC. Sanger
sequencing results and cell survival analyses demonstrated that optimal
correction
efficiencies and cell survivals were achieved with a Cas9-36GFP: sgRNA: ssODN
molar
ratio of 1:1:35:1.15 (Fig. 17).
Colony analysis for sickle correction in human iPSC
Human sickle iPSC were nucleoporated with TAT-Cas9WT-36GFP-INF7:T2
sgRNA:ssODN at a molar ratio of 1.0:1.35:1.0 to investigate the correction
efficiency in cell
populations (Fig. 18) and, subsequently, at a single cell level (Table 3). For
single cell
analysis, nucleoporated iPSCs were plated in a 96-well plate after serial
dilution. Two weeks
later, single iPSC colonies were picked, genomic DNA isolated, and Sanger
sequencing
performed. Forty-three single iPSC colonies were analyzed for on-target
modifications.
Table 3 summarizes the Sanger sequencing results for these iPSC clones. Twenty-
eight of
the 43 colonies contained at least one corrected allele (A/A, A/S or A/indel);
therefore, 65.1%
of the clones contained at least one corrected allele. iPSC containing at
least one corrected
allele will produce red blood cells that do not sickle.
46

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
Table 3. Summary for Sanger sequencing results of iPSC colonies corrected by
EpcCas9 RNP/ssODN
Total single colonies 43
A/A 14 32.6%
A/5 4 93%
S/S 3 7.0%
A/indel 10 23.3%
5/indel 6 14.0%
Indel/indel 6 14.0%
Colonies with at least 1 allele corrected 28 65.1%
Colonies with indels 22 51.2%
Colonies with genome modification 40 93.0%
Total number of alleles 86
Total "A" alleles (corrected) 42 48.8%
Total "S" alleles (uncorrected) 16 18.6%
Total "indel" alleles 28 32.6%
Genome-editing events were also assessed at the allele level for these iPSC
clones.
Forty-two of 86 alleles (48.8%) were corrected, 28 of 86 alleles (32.6%)
contained indels and
16 of 86 alleles (18.6%) were unmodified. This high rate of genome
modification (81.4% of
alleles and 93% of cells) demonstrates highly efficient gene targeting with
the biochemical
complex is possible.
Correction of human iPSC with EpcCas9 RNPs and wobble ssODNs
Retargeting of corrected DNA is a potential pitfall for the CRISPR/Cas system
in
HDR based gene correction. Compared to plasmid or viral delivery, the risk of
retargeting for
Cas9 RNP is low due to the RNPs short half-life; however, retargeting is
difficult to avoid
completely. In this example, the sickle mutation is located within the T2
sgRNA targeting
sequence and is only 2 base pairs from the PAM. After correction with the
ssODN, the
corrected DNA contains a 1 base mismatch with the sgRNA target sequence. This
difference
reduces but does not eliminate retargeting. One strategy to prevent
retargeting is to introduce
wobble base changes into the correction template. These base changes do not
alter the
translated protein sequence but alter the DNA sequences at or near the PAM
sequence so that
the corrected DNA will no longer be a target for the Cas9 RNP. Based on this
strategy, sickle
iPSC were nucleoporated with TAT-Cas9WT-36GFP-INF7/T1sgRNA/T1wb-ssODN and
TAT-Cas9WT-36GFP-INF7/T2sgRNA/T2wb-ssODN to determine whether EpcCas9 RNP
could correct the sickle mutation at high efficiencies with wobble ssODNs.
Sanger sequencing results for the nucleoporated cell populations verified
correction of
the sickle mutation in both populations of nucleoporated cells (Fig. 19). The
sickle correction
47

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
efficiency with T2wb-ssODN (Fig. 19B) was similar to the correction efficiency
of the
ssODN without wobble bases (Fig 18). However, the sickle correction efficiency
with Ti
sgRNA and Tlwb ssODN is lower than T2wb-ssODN, probably due to differences in
sgRNA
targeting efficiencies, distance from the sickle mutation to the sgRNA
cleavage sites and the
number of wobble bases. Therefore, T2wb-ssODN is the preferred ssODN.
Whole Genome Sequencing analysis of EpcCas9 corrected iPSC colonies
To determine the specificity of EpcCas9 RNP directed correction of human
sickle
patient iPSCs, Whole Genome Sequencing (WGS) was performed on uncorrected
sickle iPSC
and 4 homozygous corrected clones wereproduced with TAT-Cas9WT-36GFP-INF7 RNP.
Within the 4 corrected iPSC clones, 2 (T2-c11 and T2-c12) were corrected with
T2 sgRNA
and the 91-nt ssODN without wobble bases; 1 clone (T1w) was corrected with T2
sgRNA
and a 95-nt T2wb ssODN and 1 clone (Tlw) was corrected with Ti sgRNA and a 90-
nt
Tlwb ssODN (Table 4). These WGS data confirmed homozygous correction of the
sickle
mutation and the absence of on-target indels in the 4 homozygous corrected
iPSC clones (Fig
20A). Analysis of 4720 potential off-target sites with homology to the Ti
sgRNA and 1476
potential off-target sites with homology to the T2 sgRNA (1-5 mismatches)
demonstrated no
off-target modifications (Fig 20B). Furthermore, analysis of the whole genome
sequence
data as described in Chang et al. (Cell Reports 12(10): 1668-77 (2015),
demonstrated no
disease-causing variants in sequences with or without homology to the sgRNAs.
Four
homozygous corrected clones were produced with TAT-Cas9WT-36GFP-INF7 RNP.
Table 4. Whole Genome Sequencing analysis of EpcCas9 corrected iPSC colonies
Clone ID Cas9 protein sgRNA sgRNA sequence Wobble donor ssODN
T1w Ti GGTCTGCCGTTACTGCCCTG Ti wobble
SEQ ID NO: 68
T2w T2 GTAACGGCAGACTTCTCCAC T2 wobble
TAT-Cas9WT- SEQ ID NO: 69
36GFP-INF7
T2-c11 T2 GTAACGGCAGACTTCTCCAC No wobble
T2-c12 T2 GTAACGGCAGACTTCTCCAC No wobble
Gene correction of sickle patient bone marrow CD34+ HSPCs
Correction of primary CD34+ HSPCs from a sickle patient followed by autologous
transplant is a powerful and simple approach for SCD gene therapy. To
determine whether
48

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
EpcCas9 RNP can also correct the sickle mutation in bone marrow progenitors,
obtained
CD34+ HSPCs were obtained from bone marrow of a consenting sickle cell
patient. Sickle
CD34+ cells were purified on anti-CD34 beads, and the cell cycle was activated
by culture
for 2 days in media with specific cytokines (SCF, TPO and FLT-3).
Subsequently, the cells
were nucleoporated with Cas9WT, Cas9-36GFP or TAT-Cas9-3xTAT plus T2 sgRNA and
ssODN. The efficiency of sickle correction was determined 6 days after
nucleoporation by
the Sanger sequencing (Fig 21A). The highest correction efficiency was
obtained with
Cas9WT; however, indel frequency was high. Although the correction efficiency
with the 2
EpcCas9 RNPs was lower than with Cas9WT, the frequency of indels was
dramatically
lower.
Correction of the sickle mutation with one EpcCas9 (Cas9-36GFP) was verified
at the
mRNA and protein levels (Fig 21B-D). After expansion of the nucleoporated
cells in human
erythroid expansion media for 10 days, RT-PCR and Sanger sequencing were
performed (Fig
21B). Approximately equal amounts of betaA and betaS mRNA were observed (peaks
are
essentially superimposed). Cells were also cultured in human erythroid
differentiation media
containing Erythropoietin (Epo) for 15-18 days. The red blood cells (RBCs)
derived from this
culture were lysed, and hemoglobins were resolved on an IEF gel (Fig 21C).
Approximately
35% of total hemoglobin was HbA (Fig. 21C), and this result was confirmed by
mass
spectrometry (Fig 21D). In vivo, RBCs containing HbA survive 5-10 times longer
than rbcs
containing only Hb S. Therefore, if about 30% of cells are corrected in the
bone marrow after
transplantation, HbA levels of 60-70% will be achieved in peripheral blood.
EpcCas9 RNPs enhance the correction/indel ratio in sickle patient bone marrow
CD34+
HSPCs
In addition to examining correction of the sickle mutation in populations of
patient
bone marrow CD34+ cells, we analyzed colonies derived from single CD34+
progenitors.
After nucleoporation with TAT-Cas9WT-36GFP-INF7, CD34+ cells were mixed with
semi-
solid MethoCult media and plated into dishes. Two weeks after plating,
colonies derived
from single cells were isolated, DNA was extracted and Sanger sequence
performed. The
colonies that we examined were BFU-E (Burst Forming Units-Erythroid), CFU-E
(Colony
Forming Units-Erythroid) and CFU-GEMM (Colony Forming Units-Granulocyte,
Erythrocyte, Monocyte, Megakaryocyte). Figure 11 illustrates typical BFU-E and
CFU-
GEMM colonies (A) and representative Sanger Sequencing results of the six
genotypes that
were obtained (B). Table 5 summarizes of the Sanger sequencing results from
95, 96, and 96
colonies (BFU-E, CFU-E and CFU-GEMM) obtained after nucleoporation of Cas9WT,
49

CA 02989831 2017-12-15
WO 2016/205703 PCT/US2016/038161
Cas9WT-36GFP, and TAT-Cas9WT-3xTAT RNPs and ssODNs, respectively. The highest
correction efficiency was obtained with Cas9WT (51.6%); however, indel/indel
frequency in
cells treated with Cas9WT was also very high (40.0%). This level of
indel/indel may result
in beta-thalassemia because these HSCs will compete effectively for a limited
number of
bone marrow niches and red blood cells derived from these HSCs cannot
synthesize HbA.
Although the correction efficiency obtained with Cas9WT-36GFP RNP was lower
(28.1%),
this level of correction is sufficient to cure the disease as discussed above,
and the frequency
of indels (8.3%) is much safer. For TAT-Cas9WT-3xTAT RNP, the correction
efficiency
(32.3%) and indel frequency (14.6%) were intermediate. The correction/indel
ratios after
nucleoporation of Cas9WT, Cas9WT-36GFP, and TAT-Cas9WT-3xTAT RNPs plus ssODNs
are 1.29 (51.6/40.0), 3.39 (28.1/8.3) and 2.21 (32.3/14.6), respectively.
Therefore, Cas9WT-
36GFP that has a correction/indel ratio of 3.39 is our preferred EpcCas9.
Table 5. Summary of the Sanger sequencing results from 95, 96, and 96 colonies
(BFU-E, CFU-E and
CFU-GEMM) obtained after nucleoporation of human sickle patient bone marrow
CD34+ HSPC with
Cas9WT, Cas9WT-36GFP, and TAT-Cas9WT-3xTAT RNPs and ssODNs, respectively.
Cas9WT RNP Cas9WT-36GFP RNP
TAT-Cas9WT-3xTAT RNP
+ ssODN + ssODN + ssODN
Total colonies 95 96 96
GEMM /BFU-E /CFU-E colonies 7/81/7 16/80/0 10/86/0
A/A 18 18.9% 3 3.1% 10
10.4%
A/5 2 2.1% 14 14.6% 4
4.2%
5/5 5 5.3% 46 47.9% 27
28.1%
A/indel 29 30.5% 10 10.4% 17 17.7%
S/indel 3 3.2% 15 15.6% 24 25.0%
Indel/indel 38 40.0% 8 8.3% 14
14.6%
Colonies with at least 1 allele corrected 49 51.6% 27 28.1%
31 32.3%
Colonies with indels 70 73.7% 41 42.7% 55
57.3%
Colonies with genome modification 90 94.7% 50 52.1% 69
71.9%
GRAM correction 5 71.4% 4 25.0% 1
10.0%
Total number of alleles 190 192 192
Total "A" alleles (corrected) 67 35.3% 30 15.6% 41
21.4%
Total "S" alleles (uncorrected) 15 7.9% 121 63.7% 82
43.2%
Total "indel" alleles 108 56.8% 41 21.6% 69
36.3%
As discussed above, the sickle correction efficiency of the Cas9WT-36GFP
RNP/ssODN complex (28.1% of total CFU; 25% of CFU-GEMM) is high enough to cure
the
disease. This level of correction in the bone marrow after transplantation
would result in 60-
70% corrected RBC in peripheral blood. In addition, only 8.3% of colonies are
homozygous
indels (indel/indel); therefore, thalassemia is unlikely to result after
transplantation.

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
EpcCas9 RNPs enhance cell survival after nucleoporation in sickle patient bone
marrow
CD34+ HSPCs
The data in Fig. 22C demonstrate that EpcCas9 RNPs enhance cell survival after
nucleoporation in sickle patient bone marrow CD34+ HSPCs. The number of
erythroid
colonies (BFU-E and CFU-E) obtained after nucleoporation of sickle patient
bone marrow
CD34+ HSPCs was compared with Cas9WT, Cas9WT-36GFP, and TAT-Cas9WT-3xTAT
RNPs plus ssODNs. The number of colonies obtained with Cas9WT RPN/ssODN was
normalized to 1. The number of colonies obtained with Cas9WT-36GFP RNP/ssODN
was
2.5-fold higher than the Cas9WT control and TAT-Cas9WT-3xTAT RNP/ssODN was 1.6-
fold higher. It was concluded that Epc (Engineered positive charge) protects
human bone
marrow progenitors/stem cells from the toxic effects of single stranded
oligodeoxynucleotides (ssODNs).
These results are significant because the dose of CD34+ HPSCs is critical for
bone
marrow reconstitution after transplantation. In general, two million CD34+
cells/kg are
transplanted into human recipients. Cell doses below this level result in poor
long-term
reconstitution. A 75kg patient requires a dose of approximately 150 million
cells. One liter
of bone marrow can be harvested from a 75kg patient under anesthesia and
approximately
200 million CD34+ cells can be isolated for transplantation. As indicated
above, 2.5-fold
fewer cells are obtained after nucleoporation of CD34+ cells with Cas9WT
RNP/ssODN
compared to Cas9-36GFP RNP/ssODN. Therefore, our preferred complex for
correction is
Cas9WT-36GFP RNP/ssODN.
EpcCas9 results in higher genome editing specificity
To evaluate the specificity of genome editing by EpcCas9 RNPs in nucleoporated
CD34+ cells, deep sequencing analysis was conducted at five potential off-
target genomic
loci. The five potential off-target sites were the top 5 sites predicted by
the Zhang MIT server
(http://crispr.mit.edu) based on sequence homology to the sgRNA. In Cas9
RNP/ssODN
nucleoporated sickle patient CD34+ cells, deep sequencing measured
approximately 0.1%
off-target indels at OT5 site (Table 6). In contrast, in Cas9WT-36GFP or TAT-
Cas9WT-
3xTAT RNP/ssODN nucleoporated cells, no off-target modifications were observed
(Fig.
23).
51

CA 02989831 2017-12-15
WO 2016/205703 PCT/US2016/038161
Table 6 Deep sequencing analysis of 5 potential off-target genomic loci to
evaluate editing
specificity of EpcCas9 RNPs in nucleoporated CD34+ cells
OT1 0T2 0T3 0T4
0T5
chr3:37684838 chr12:112746615 chr11:132762118 chr14:101366447 chr10:95158973
3MMs [1:5:7] 3MMs [2:4:11] 3MMs
[2:5:19] 4MMs [1:2:5:7] 4MMs [1:2:3:7]
Indel reads 13 34 28 4
8
N Non-indel reads 209990 900262 700844
449423 226882
eg ctr l
Total reads 210003 900296 700872 449427
226890
Indel percentage 0.0062% 0.0038% 0.0040% 0.0009%
0.0035%
Indel reads 6 33 37 7
240
9WT Non-indel reads 199453 862095 754410 425039 226916
Cas
Total reads 199459 862128 754447 425046
227156
Indel percentage 0.0030% 0.0038% 0.0049% 0.0016%
0.1057%
Indel reads 4 37 23 4
8
Non-indel reads 189683 777630 615613 482843
207613
Cas9WT-36GFP
Total reads 189687 777667 615636 482847
207621
Indel percentage 0.0021% 0.0048% 0.0037% 0.0008%
0.0039%
Indel reads 9 24 32 3
9
Non-indel reads 193690 843834 685044 458625
199515
TAT-Cas9WT-3xTAT
Total reads 193699 843858 685076 458628
199524
Indel percentage 0.0046% 0.0028% 0.0047% 0.0007%
0.0045%
In addition, in erythroid colonies derived from Cas9WT RNP/ssODN nucleoporated
sickle CD34+ cells, 5 out of 95 colonies containing non-specific modifications
near
(upstream or downstream) the targeting site were observed (Fig. 24). These non-
specific
modifications are random gene replacements or indels that do not appear to be
initiated at the
expected Cas9 RNP cutting site. In contrast, 0 out of 96 colonies derived from
EpcCas9
RNPs nucleoporated cells contain non-specific modifications.
Example 4
Correction of Sickle Cell Mutation in mice
Figure 25 shows an isoelectric focusing (IEF) gel analysis of blood six weeks
after
primary transplantation of Sickle Mouse Fetal Liver c-Kit+ cells nucleoporated
with Cas9
RNP/ssODN to correct a sickle cell mutation. Mouse fetal liver c-kit+ cells
are equivalent to
human cord-blood Cd34+ cells. Figure 26 shows ddPCR analysis of FACS purified
bone
marrow cells at twelve weeks post-transplantation into irradiated C57B16 mice.
Twelve
weeks after nucleoporation and transplantation, approximately 50% of erythroid
cells
(Ten 19+) and myeloid cells (CD11b+ and CD11b+/GR1+) are corrected. Erythroid
and
myeloid cells are relatively short lived; therefore, these cells are derived
from transplanted
HSCs. Correction levels in B and T cells should rise to approximately 50%
after secondary
transplantation at twelve weeks (twenty-four weeks total). After twenty-four
weeks, most if
52

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
not all hematopoietic cells will be derived from long-term HSCs. Figure 27
shows IEF gel
analysis of the blood in mice twelve weeks after primary transplantation and
six weeks after
secondary transplantation of cells nucleoporated with Cas9 RNP/ssODN to
correct a sickle
cell mutation. Human HbA is produced in mice after transplantation of HSCs
nucleoporated
with Cas9 RNP/ssODN to correct a sickle cell mutation. The mouse hemoglobin
band will
disappear in six more weeks.
Sequences
SEQ ID NO: 1
TAACGGCAGACTTCTCCAC
SEQ ID NO: 2
GTAAC GGC AGAC T TC TC CAC GT TTTAGAGC TAGAAATAGC AAGTTAAAATAAGG
CTAGTCCGTTATCAACTTGAAAAAGTGGCACCGAGTCGGTGCTTTTTTT
SEQ ID NO:3
Cas9-supercharged GFP construct
m dykdhdgdykdhdi dykdd ddkm apkkkrkvgi hgvp aadkky si gl di gtn svgwavitdeykvp
skkfkvl gntdrh
sikknligallfdsgetaeatrlkrtarrrytrrknricylqeifsnemakvddsffhrleesflveedkkherhpifg
nivdevayhek
yptiyhlrkklvdstdkadlrliyl al ahmi kfrghfl i egdl np dn sdvdkl fi qlvqtynql
feenpi nasgvdakailsarl sksrrl
enli aqlpgekknglfgnli al slgltpnflcsnfdlaedaklql
skdtydddldnllaqigdqyadlflaaknl sdaill sdilrvnteitk
apl sasmikrydehhqdltllkalvrqqlpekykeiffdqskngyagyi
dggasqeefykfikpilekmdgteellvklnredllrk
qrtfdng si phqi hl gel hailrrqe dfypfl kdnreki eki ltfri pyyvgpl argn
srfawmtrkseetitpwnfeevvdkgas aq s
fi ermtnfdknl pnekvl pkh sl lyeyftvyneltkvkyvtegmrkp afl
sgeqkkaivdllfktnrkvtvkqlkedyfkkiecfds
vei
sgvedrfnaslgtyhdllkiikdkdfldneenediledivltltlfedremieerlktyahlfddkvmkqlkrrrytgw
grl srkli
ngi rdkq sgktildfl ks dgfanrnfm ql i hdd sltfke di qkaqvsgqgd sl hehi anl agsp
ai kkgilqtvkvvdelvkvm gr
hkp enivi em arenqttqkgqkn srermkri e egi kel g sqilkehpventql qneklylyyl
qngrdmyvdqel di nrl sdydv
dhivpqsflkddsidnkvltrsdknrgksdnvpseevykkmknywrqllnaklitqrkfdnitkaerggl
seldkagfikrqlvetr
qitkhvaqildsrmntkydendklirevkvitlksklvsdfrkdfqfykvreinnyhhahdaylnavvgtalikkypkl
esefvyg
dykvy dvrkmi aks eqei gkatakyffy sni mnffkteitl angeirkrpl i
etngetgeivwdkgrdfatvrkvl smpqvnivkk
tevqtggfskesilpkrnsdkliarkkdwdpkkyggfdsptvaysylvvakvekgkskklksvkellgitimerssfek
npidfle
akgykevkkdl i i kl pky sl fel engrkrml a sagel qkgnel al p skyvnflyl ashyekl
kgsp edne qkql fve qhkhyl dei
ieqi sefskrviladanldkvl s aynkhrdkpi reqaeni i hlftltnl gap aafkyfdtti
drkrytstkevl datl i hq sitgly etri dl s
qlggdkrpaatkkagqakkkkgsgsngssgsaskgerlfrgkvpilvelkgdvnghkfsvrgkgkgdatrgkltlkfic
ttgklpv
pwpthrttltygvqcfsrypkhmkrhdffksampkgyvqerti
sfkkdgkyktraevkfegrtlynriklkgrdfkekgnilghk1
rynfn shkvyitadkrkngi kakfki rhnvkdg svql adhy qqntpi grgpvllprnhyl strskl
skdpkekrdhmvllefvtaa
gikhgrderyk
53

CA 02989831 2017-12-15
WO 2016/205703
PCT/US2016/038161
SEQ ID NO: 4
TAT-Cas9-supercharged GFP construct
ygrkkrrqrrrppqaggsmdykdhdgdykdhdi dykddddkm apkkkrkvgi hgvp aadkky si gl di
gtnsvgwavitd
eykvp skkfkvlgntdrhsikknligallfdsgetaeatrlkrtarrrytrrknri
cylqeifsnemakvddsffhrlee sflveedkkh
erhpi fgnivdevayhekyptiyhl rkklvd stdkadl rl iyl al ahmi kfrghfl i egdl np dn
s dvdkl fi qlvqtynqlfeenpin
a sgvdakail sari sksrrl enl i aql pgekkngl fgnl i al sl gltpnfksnfdl ae dakl
ql skdtydddldnllaqigdqyadlflaa
knl sdaill sdilrvnteitkapl sasmikrydehhqdltllkalvrqqlpekykeiffdqskngyagyi
dggasqeefykfikpil ek
m dgte el lvkl nre dllrkqrtfdng si phqi hl gel hailrrqedfypfl kdnreki eki
ltfri pyyvgpl argn srfawmtrkseet
itpwnfeevvdkgasaqsfi ermtnfdknl pnekvl pkh sl ly eyftvyneltkvkyvtegmrkp afl
sgeqkkaivdllfktnrk
vtvkqlkedyfkki ecfdsvei sgvedrfnaslgtyhdllkiikdkdfldneenediledivltltlfedremi
eerlktyahlfddkv
mkqlkrrrytgwgrl srkl i ngi rdkq sgktildfl ks dgfanrnfmql i hdd sltfke di
qkaqvsgqgd sl hehi anl agsp ai kk
gilqtvkvvdelvkvmgrhkpenivi emarenqttqkgqknsrermkri
eegikelgsqilkehpventqlqneklylyylqngr
dmyvdqel di nrl sdydvdhivpqsflkddsi dnkvltrsdknrgksdnvp se evykkmknywrqllnakl
itqrkfdnitkaer
ggl sel dkagfi krqlvetrqitkhvaqild srmntky dendkl i revkvitl ksklvs
dfrkdfqfykvrei nnyhhahdayl navy
gtalikkypkl e sefvygdykvy dvrkmi akse qei gkatakyffy sni mnffkteitl angei
rkrpl i etngetgeivwdkgrdf
atvrkvl
smpqvnivkktevqtggfskesilpkrnsdkliarkkdwdpkkyggfdsptvaysylvvakvekgkskklksvkell
gin m ers sfeknpi dfl eakgykevkkdl i i kl pky sl fel engrkrml a sagel qkgnel
al p skyvnflylashyeklkgsped
neqkqlfveqhkhyldeii eqi sefskrviladanldkvl s aynkhrdkpi reqaeni i hlftltnl gap
aafkyfdtti drkrytstke
vldatlihqsitglyetri dl
sqlggdkrpaatkkagqakkkkgsgsngssgsaskgerlfrgkvpilvelkgdvnghkfsvrgkgk
gdatrgkltlkfi cttgkl pvpwptivttltygvqcfsrypkhmkrh dffksampkgyvqerti
sfkkdgkyktraevkfegrtivnr
iklkgrdfkekgnilghklrynfnshkvyitadkrkngikakfkirhnvkdgsvq1adhyqqntpigrgpvllprnhyl
strskl sk
dpkekrdhmvllefvtaagikhgrderykggsggsvdglfeai egfiengwegmi dgwyg
54

Representative Drawing
A single figure which represents the drawing illustrating the invention.
Administrative Status

2024-08-01:As part of the Next Generation Patents (NGP) transition, the Canadian Patents Database (CPD) now contains a more detailed Event History, which replicates the Event Log of our new back-office solution.

Please note that "Inactive:" events refers to events no longer in use in our new back-office solution.

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Event History , Maintenance Fee  and Payment History  should be consulted.

Event History

Description Date
Application Not Reinstated by Deadline 2022-09-08
Inactive: Dead - RFE never made 2022-09-08
Deemed Abandoned - Failure to Respond to a Request for Examination Notice 2021-09-08
Letter Sent 2021-06-17
Maintenance Fee Payment Determined Compliant 2020-12-14
Common Representative Appointed 2020-11-07
Letter Sent 2020-08-31
Inactive: COVID 19 - Deadline extended 2020-08-19
Inactive: COVID 19 - Deadline extended 2020-08-06
Inactive: COVID 19 - Deadline extended 2020-07-16
Inactive: COVID 19 - Deadline extended 2020-07-02
Inactive: COVID 19 - Deadline extended 2020-06-10
Common Representative Appointed 2019-10-30
Common Representative Appointed 2019-10-30
Change of Address or Method of Correspondence Request Received 2019-07-24
Inactive: Delete abandonment 2018-09-17
Inactive: Office letter 2018-09-17
Inactive: Correspondence - MF 2018-08-30
Inactive: Correspondence - PCT 2018-08-03
Deemed Abandoned - Failure to Respond to Maintenance Fee Notice 2018-06-18
Inactive: Cover page published 2018-03-02
Inactive: Notice - National entry - No RFE 2018-01-10
Inactive: IPC assigned 2018-01-03
Inactive: IPC assigned 2018-01-03
Inactive: IPC assigned 2018-01-03
Inactive: IPC assigned 2018-01-03
Inactive: IPC assigned 2018-01-03
Inactive: IPC assigned 2018-01-03
Application Received - PCT 2018-01-03
Inactive: First IPC assigned 2018-01-03
Letter Sent 2018-01-03
Letter Sent 2018-01-03
National Entry Requirements Determined Compliant 2017-12-15
BSL Verified - No Defects 2017-12-15
Inactive: Sequence listing - Received 2017-12-15
Inactive: Sequence listing - Received 2017-12-15
Application Published (Open to Public Inspection) 2016-12-22

Abandonment History

Abandonment Date Reason Reinstatement Date
2021-09-08
2018-06-18

Maintenance Fee

The last payment was received on 2022-06-10

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Fee History

Fee Type Anniversary Year Due Date Paid Date
MF (application, 2nd anniv.) - standard 02 2018-06-18 2017-12-15
Basic national fee - standard 2017-12-15
Registration of a document 2017-12-15
MF (application, 3rd anniv.) - standard 03 2019-06-17 2019-06-05
Late fee (ss. 27.1(2) of the Act) 2020-12-14 2020-12-14
MF (application, 4th anniv.) - standard 04 2020-08-31 2020-12-14
MF (application, 5th anniv.) - standard 05 2021-06-17 2021-06-11
MF (application, 6th anniv.) - standard 06 2022-06-17 2022-06-10
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
THE UAB RESEARCH FOUNDATION
Past Owners on Record
CHIA-WEI CHANG
LEI DING
TIM TOWNES
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column (Temporarily unavailable). To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2017-12-14 54 3,387
Drawings 2017-12-14 29 2,259
Claims 2017-12-14 9 311
Abstract 2017-12-14 2 98
Representative drawing 2017-12-14 1 38
Cover Page 2018-03-01 1 65
Courtesy - Certificate of registration (related document(s)) 2018-01-02 1 106
Courtesy - Certificate of registration (related document(s)) 2018-01-02 1 106
Notice of National Entry 2018-01-09 1 193
Reminder of maintenance fee due 2018-02-19 1 111
Commissioner's Notice - Maintenance Fee for a Patent Application Not Paid 2020-10-12 1 537
Courtesy - Acknowledgement of Payment of Maintenance Fee and Late Fee 2020-12-13 1 432
Commissioner's Notice: Request for Examination Not Made 2021-07-07 1 541
Courtesy - Abandonment Letter (Request for Examination) 2021-09-28 1 552
PCT Correspondence 2018-08-02 3 209
Maintenance fee correspondence 2018-08-29 3 215
Courtesy - Office Letter 2018-09-16 1 48
National entry request 2017-12-14 22 852
Patent cooperation treaty (PCT) 2017-12-14 2 97
Declaration 2017-12-14 6 112
International search report 2017-12-14 4 118
Maintenance fee payment 2020-12-13 1 30

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :